1
|
Mo W, Donahue JK. Gene therapy for atrial fibrillation. J Mol Cell Cardiol 2024; 196:84-93. [PMID: 39270930 PMCID: PMC11534567 DOI: 10.1016/j.yjmcc.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in adults. Current limitations of pharmacological and ablative therapies motivate the development of novel therapies as next generation treatments for AF. The arrhythmia mechanisms creating and sustaining AF are key elements in the development of this novel treatment. Gene therapy provides a useful platform that allows us to regulate the mechanisms of interest using a suitable transgene(s), vector, and delivery method. Effective gene therapy strategies in the literature have targeted maladaptive electrical or structural remodeling that increase vulnerability to AF. In this review, we will summarize key elements of gene therapy for AF, including molecular targets, gene transfer vectors, atrial gene delivery and preclinical efficacy and toxicity testing. Recent advances and challenges in the field will be also discussed.
Collapse
Affiliation(s)
- Weilan Mo
- From the Division of Cardiology, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - J Kevin Donahue
- From the Division of Cardiology, University of Massachusetts Medical School, Worcester, MA, United States of America.
| |
Collapse
|
2
|
Wijdeveld LFJM, Collinet ACT, Huiskes FG, Brundel BJJM. Metabolomics in atrial fibrillation - A review and meta-analysis of blood, tissue and animal models. J Mol Cell Cardiol 2024; 197:108-124. [PMID: 39476947 DOI: 10.1016/j.yjmcc.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/03/2024] [Accepted: 10/18/2024] [Indexed: 11/10/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) is a highly prevalent cardiac arrhythmia associated with severe cardiovascular complications. AF presents a growing global challenge, however, current treatment strategies for AF do not address the underlying pathophysiology. To advance diagnosis and treatment of AF, a deeper understanding of AF root causes is needed. Metabolomics is a fast approach to identify, quantify and analyze metabolites in a given sample, such as human serum or atrial tissue. In the past two decades, metabolomics have enabled research on metabolite biomarkers to predict AF, metabolic features of AF, and testing metabolic mechanisms of AF in animal models. Due to the field's rapid evolution, the methods of AF metabolomics studies have not always been optimal. Metabolomics research has lacked standardization and requires expertise to face methodological challenges. PURPOSE OF THE REVIEW We summarize and meta-analyze metabolomics research on AF in human plasma and serum, atrial tissue, and animal models. We present the current progress on metabolic biomarkers candidates, metabolic features of clinical AF, and the translation of metabolomics findings from animal to human. We additionally discuss strengths and weaknesses of the metabolomics method and highlight opportunities for future AF metabolomics research.
Collapse
Affiliation(s)
- Leonoor F J M Wijdeveld
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, the Netherlands; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, MA 02142, Cambridge, United States
| | - Amelie C T Collinet
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, the Netherlands
| | - Fabries G Huiskes
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, the Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Bode D, Pronto JRD, Schiattarella GG, Voigt N. Metabolic remodelling in atrial fibrillation: manifestations, mechanisms and clinical implications. Nat Rev Cardiol 2024; 21:682-700. [PMID: 38816507 DOI: 10.1038/s41569-024-01038-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/01/2024]
Abstract
Atrial fibrillation (AF) is a continually growing health-care burden that often presents together with metabolic disorders, including diabetes mellitus and obesity. Current treatments often fall short of preventing AF and its adverse outcomes. Accumulating evidence suggests that metabolic disturbances can promote the development of AF through structural and electrophysiological remodelling, but the underlying mechanisms that predispose an individual to AF are aetiology-dependent, thus emphasizing the need for tailored therapeutic strategies to treat AF that target an individual's metabolic profile. AF itself can induce changes in glucose, lipid and ketone metabolism, mitochondrial function and myofibrillar energetics (as part of a process referred to as 'metabolic remodelling'), which can all contribute to atrial dysfunction. In this Review, we discuss our current understanding of AF in the setting of metabolic disorders, as well as changes in atrial metabolism that are relevant to the development of AF. We also describe the potential of available and emerging treatment strategies to target metabolic remodelling in the setting of AF and highlight key questions and challenges that need to be addressed to improve outcomes in these patients.
Collapse
Affiliation(s)
- David Bode
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
4
|
Cousineau JP, Dawe AM, Alpaugh M. Investigating the Interplay between Cardiovascular and Neurodegenerative Disease. BIOLOGY 2024; 13:764. [PMID: 39452073 PMCID: PMC11505144 DOI: 10.3390/biology13100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024]
Abstract
Neurological diseases, including neurodegenerative diseases (NDDs), are the primary cause of disability worldwide and the second leading cause of death. The chronic nature of these conditions and the lack of disease-modifying therapies highlight the urgent need for developing effective therapies. To accomplish this, effective models of NDDs are required to increase our understanding of underlying pathophysiology and for evaluating treatment efficacy. Traditionally, models of NDDs have focused on the central nervous system (CNS). However, evidence points to a relationship between systemic factors and the development of NDDs. Cardiovascular disease and related risk factors have been shown to modify the cerebral vasculature and the risk of developing Alzheimer's disease. These findings, combined with reports of changes to vascular density and blood-brain barrier integrity in other NDDs, such as Huntington's disease and Parkinson's disease, suggest that cardiovascular health may be predictive of brain function. To evaluate this, we explore evidence for disruptions to the circulatory system in murine models of NDDs, evidence of disruptions to the CNS in cardiovascular disease models and summarize models combining cardiovascular disruption with models of NDDs. In this study, we aim to increase our understanding of cardiovascular disease and neurodegeneration interactions across multiple disease states and evaluate the utility of combining model systems.
Collapse
Affiliation(s)
| | | | - Melanie Alpaugh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (J.P.C.); (A.M.D.)
| |
Collapse
|
5
|
Xu B, Xu Y, Ren W, Meng S, Hong T, Cao Z, Xiao X, Guo X, Yu L, Zhao J, Wang H. S-Methyl-L-cysteine targeting MsrA attenuates Ang II-induced oxidative stress and atrial remodeling via the p38 MAPK signaling pathway. Food Funct 2024; 15:9165-9175. [PMID: 39157962 DOI: 10.1039/d4fo03078h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Atrial fibrillation (AF) is the most prevalent sustained tachyarrhythmia in patients with cardiovascular diseases. Recently, it has been discovered that oxidative stress is an important contributor to AF. Therefore, antioxidant therapies for AF have great potential for clinical applications. Methionine, a sulfur-containing amino acid residue other than cysteine, is recognized as a functional redox switch, which could be rescued from the reversible oxidation of methionine sulfoxide by methionine sulfoxide reductase A (MsrA). S-Methyl-L-cysteine (SMLC), a natural analogue of Met, which is abundantly found in garlic and cabbage, could substitute for Met oxidations and mediate MsrA to scavenge free radicals. However, whether SMLC alleviates AF is unclear. This study aims to clarify the effects of SMLC on AF and elucidate the underlying pharmacological and molecular mechanisms. In vivo, SMLC (70, 140 and 280 mg kg-1 day-1) was orally administered to mice for 4 weeks with angiotensin II (Ang II) by subcutaneous infusion using osmotic pumps to induce AF. Ang II significantly prompted high AF susceptibility and atrial remodeling characterized by oxidative stress, conductive dysfunction and fibrosis. SMLC played a remarkable protective role in Ang II-induced atrial remodeling dose-dependently. Moreover, RNA sequencing was performed on atrial tissues to identify the differentially expressed mRNA, which was to screen out MSRA, CAMK2 and MAPK signaling pathways. Western blots confirmed that Ang II-induced downregulation of MsrA and upregulation of oxidized CaMKII (ox-CaMKII) and p38 MAPK could be reversed in a concentration-dependent manner by SMLC. To investigate the underlying mechanisms, HL-1 cells (mouse atria-derived cardiomyocytes) treated with Ang II were used for an in vitro model. SMLC alleviated Ang II-induced cytotoxicity, mitochondrial damage and oxidative stress. Additionally, knockdown MsrA could attenuate the protective effects of SMLC, which were eliminated by the p38 MAPK inhibitor SB203580. In summary, the present study demonstrates that SMLC protects against atrial remodeling in AF by inhibiting oxidative stress through the mediation of the MsrA/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Beibei Xu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
- Postgraduate College, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinli Xu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
| | - Wenpu Ren
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
- Postgraduate College, Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Shan Meng
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
- Postgraduate College, Jinzhou Medical University, Jinzhou 121001, China
| | - Tao Hong
- Postgraduate College, Dalian Medical University, Dalian 116044, China
- Pediatric Surgery Ward, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518052, China
| | - Zijun Cao
- Postgraduate College, Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Xiong Xiao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
- Postgraduate College, China Medical University, Shenyang 110122, China
| | - Xiaodong Guo
- Postgraduate College, Dalian Medical University, Dalian 116044, China
| | - Liming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
| | - Jikai Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
| | - Huishan Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
| |
Collapse
|
6
|
Kambayashi R, Goto A, Takahara A, Saito H, Izumi-Nakaseko H, Takei Y, Akie Y, Hori M, Sugiyama A. Characterization of remodeling processes in the atria of atrioventricular block dogs: Utility as an early-stage atrial fibrillation model. J Pharmacol Sci 2024; 156:19-29. [PMID: 39068031 DOI: 10.1016/j.jphs.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/08/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
To characterize utility of atrioventricular block (AVB) dogs as atrial fibrillation (AF) model, we studied remodeling processes occurring in their atria in acute (<2 weeks) and chronic (>4 weeks) phases. Fifty beagle dogs were used. Holter electrocardiogram demonstrated that paroxysmal AF occurred immediately after the production of AVB, of which duration tended to be prolonged in chronic phase. Electrophysiological analysis showed that inter-atrial conduction time and duration of burst pacing-induced AF increased in the chronic phase compared with those in the acute phase, but that atrial effective refractory period was hardly altered. Echocardiographic study revealed that diameters of left atrium, right pulmonary vein and inferior vena cava increased similarly in the acute and chronic phases. Histological evaluation indicated that hypertrophy and fibrosis in atrial tissue increased in the chronic phase. Electropharmacological characterization showed that i.v. pilsicainide effectively suppressed burst pacing-induced AF with increasing atrial conduction time and refractoriness of AVB dogs in chronic phase, but that i.v. amiodarone did not exert such electrophysiological effects. Taken together, AVB dogs in chronic phase appear to possess such pathophysiology as developed in the atria of early-stage AF patients, and therefore they can be used to evaluate drug candidates against early-stage AF.
Collapse
Affiliation(s)
- Ryuichi Kambayashi
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - Ai Goto
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - Akira Takahara
- Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama, Funabashi-shi, Chiba 274-8510, Japan
| | - Hiroyuki Saito
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan; CMIC Bioresearch Center, CMIC Pharma Science Co., Ltd., 10221 Kobuchisawa, Hokuto, Yamanashi 408-0044, Japan
| | - Hiroko Izumi-Nakaseko
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - Yoshinori Takei
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - Yasuki Akie
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan; CMIC Bioresearch Center, CMIC Pharma Science Co., Ltd., 10221 Kobuchisawa, Hokuto, Yamanashi 408-0044, Japan
| | - Masaaki Hori
- Department of Radiology, Faculty of Medicine, Toho University, 6-11-1 Omori-nishi, Ota-ku, Tokyo 143-8541, Japan
| | - Atsushi Sugiyama
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan; Yamanashi Research Center of Clinical Pharmacology, 73-5 Hatta, Fuefuki, Yamanashi 406-0023, Japan; Organization for the Promotion of Research and Social Collaboration, University of Yamanashi, 4-4-37 Takeda, Kofu, Yamanashi 400-8510, Japan.
| |
Collapse
|
7
|
Shen D, Zhu Y, Mao J, Lin R, Jiang X, Liang L, Peng J, Cao Y, Dong S, He K, Wang N. Highly sensitive and accurate measurement of underivatized phosphoenolpyruvate in plasma and serum via EDTA-facilitated hydrophilic interaction liquid chromatography-tandem mass spectrometry. Talanta 2024; 275:126134. [PMID: 38692044 DOI: 10.1016/j.talanta.2024.126134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024]
Abstract
Phosphoenolpyruvate (PEP) is an essential intermediate metabolite that is involved in various vital biochemical reactions. However, achieving the direct and accurate quantification of PEP in plasma or serum poses a significant challenge owing to its strong polarity and metal affinity. In this study, a sensitive method for the direct determination of PEP in plasma and serum based on ethylenediaminetetraacetic acid (EDTA)-facilitated hydrophilic interaction liquid chromatography-tandem mass spectrometry was developed. Superior chromatographic retention and peak shapes were achieved using a zwitterionic stationary-phase HILIC column with a metal-inert inner surface. Efficient dechelation of PEP-metal complexes in serum/plasma samples was achieved through the introduction of EDTA, resulting in a significant enhancement of the PEP signal. A PEP isotopically labelled standard was employed as a surrogate analyte for the determination of endogenous PEP, and validation assessments proved the sensitivity, selectivity, and reproducibility of this method. The method was applied to the comparative quantification of PEP in plasma and serum samples from mice and rats, as well as in HepG2 cells, HEK293T cells, and erythrocytes; the results confirmed its applicability in PEP-related biomedical research. The developed method can quantify PEP in diverse biological matrices, providing a feasible opportunity to investigate the role of PEP in relevant biomedical research.
Collapse
Affiliation(s)
- Danning Shen
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Yingjie Zhu
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Jie Mao
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Runfeng Lin
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Xin Jiang
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Longhui Liang
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Jing Peng
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Yanqing Cao
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Suhe Dong
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Kun He
- National Center of Biomedical Analysis, Beijing, 100850, China.
| | - Na Wang
- National Center of Biomedical Analysis, Beijing, 100850, China.
| |
Collapse
|
8
|
Cizauskas HE, Burnham HV, Panni A, Peña A, Alvarez-Arce A, Davis MT, Araujo KN, Delligatti CE, Edassery S, Kirk JA, Arora R, Barefield DY. Proteolytic degradation of atrial sarcomere proteins underlies contractile defects in atrial fibrillation. Am J Physiol Heart Circ Physiol 2024; 327:H460-H472. [PMID: 38940916 PMCID: PMC11442024 DOI: 10.1152/ajpheart.00148.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024]
Abstract
Atrial fibrillation (AFib) is the most common cardiac rhythm disturbance, often treated via electrical cardioversion. Following rhythm restoration, a period of depressed mechanical function known as atrial stunning occurs, suggesting that defects in contractility occur in AFib and are revealed upon restoration of rhythm. This project aims to define the contractile remodeling that occurs in AFib. To assess contractile function, we used a canine atrial tachypacing model of induced AFib. Mass spectrometry analysis showed dysregulation of contractile proteins in samples from AFib compared with sinus rhythm atria. Atrial cardiomyocytes show reduced force of contraction, decreased resting tension, and increased calcium sensitivity in skinned single cardiomyocyte studies. These alterations correlated with degradation of myofilament proteins including myosin heavy chain altering force of contraction, titin altering resting tension, and troponin I altering calcium sensitivity. We measured degradation of other myofilament proteins, including cardiac myosin binding protein C and actinin, that show degradation products in the AFib samples that are absent in the sinus rhythm atria. Many of the degradation products appeared as discrete cleavage products that are generated by calpain proteolysis. We assessed calpain activity and found it to be significantly increased. These results provide an understanding of the contractile remodeling that occurs in AFib and provide insight into the molecular explanation for atrial stunning and the increased risk of atrial thrombus and stroke in AFib.NEW & NOTEWORTHY Atrial fibrillation is the most common cardiac rhythm disorder, and remodeling during atrial fibrillation is highly variable between patients. This study has defined the biophysical changes in contractility that occur in atrial fibrillation along with identifying potential molecular mechanisms that may drive this remodeling. This includes proteolysis of several myofilament proteins including titin, troponin I, myosin heavy chain, myosin binding protein C, and actinin, which is consistent with the observed contractile deficits.
Collapse
Affiliation(s)
- Hannah E Cizauskas
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Hope V Burnham
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Azaria Panni
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Alexandra Peña
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Alejandro Alvarez-Arce
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - M Therese Davis
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Kelly N Araujo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Christine E Delligatti
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Seby Edassery
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Rishi Arora
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - David Y Barefield
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| |
Collapse
|
9
|
Aromolaran KA, Corbin A, Aromolaran AS. Obesity Arrhythmias: Role of IL-6 Trans-Signaling. Int J Mol Sci 2024; 25:8407. [PMID: 39125976 PMCID: PMC11313575 DOI: 10.3390/ijms25158407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Obesity is a chronic disease that is rapidly increasing in prevalence and affects more than 600 million adults worldwide, and this figure is estimated to increase by at least double by 2030. In the United States, more than one-third of the adult population is either overweight or obese. The global obesity epidemic is a major risk factor for the development of life-threatening arrhythmias occurring in patients with long QT, particularly in conditions where multiple heart-rate-corrected QT-interval-prolonging mechanisms are simultaneously present. In obesity, excess dietary fat in adipose tissue stimulates the release of immunomodulatory cytokines such as interleukin (IL)-6, leading to a state of chronic inflammation in patients. Over the last decade, increasing evidence has been found to support IL-6 signaling as a powerful predictor of the severity of heart diseases and increased risk for ventricular arrhythmias. IL-6's pro-inflammatory effects are mediated via trans-signaling and may represent a novel arrhythmogenic risk factor in obese hearts. The first selective inhibitor of IL-6 trans-signaling, olamkicept, has shown encouraging results in phase II clinical studies for inflammatory bowel disease. Nevertheless, the connection between IL-6 trans-signaling and obesity-linked ventricular arrhythmias remains unexplored. Therefore, understanding how IL-6 trans-signaling elicits a cellular pro-arrhythmic phenotype and its use as an anti-arrhythmic target in a model of obesity remain unmet clinical needs.
Collapse
Affiliation(s)
- Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
| | - Andrea Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
10
|
Cui M, Wang W, Han X, Lu Z, Yang X, Liu L, Zhou X, Chen S, Wei L, Chen N, He C, Yang G. Designing Microneedle Patch for Prophylaxis of Postoperative Atrial Fibrillation. ACS NANO 2024; 18:18889-18899. [PMID: 39004829 DOI: 10.1021/acsnano.4c00528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Postoperative atrial fibrillation (POAF) is a common complication following cardiac surgery, which often occurs within 30 postoperative days, especially peaking at 2-3 days. Antiarrhythmic medications such as amiodarone are recommended in clinical practice for the prophylaxis and treatment of POAF. However, conventional oral administration is hindered due to delayed drug action and high risks of systemic toxicity, and emerging localized delivery strategies suffer from a limited release duration (less than 30 days). Herein, we develop a microneedle (MN) patch for localized delivery of amiodarone to the atria in a "First Rapid and Then Sustained" dual-release mode. Specifically, this patch is composed of a needle array integrated with an amiodarone-loaded reservoir for a sustained and steady release for over 30 days; and an amiodarone-containing coating film deposited on the needle surface via the Langmuir-Blodgett technique for a rapid release at the first day. Upon this design, only one MN patch enables a higher drug accumulation in the atrial tissue at the first day than oral administration and simultaneously remains therapeutical levels for over 30 days, despite at a significantly reduced drug dosage (5.08 mg in total versus ∼10 mg per day), thereby achieving ideal preventive effects and safety in a rat model. Our findings indicate that this MN device provides a robust and efficient delivery platform for long-term prophylaxis of POAF.
Collapse
Affiliation(s)
- Mingrui Cui
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Wenshuo Wang
- Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Xiaoyue Han
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Ziyi Lu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xuexia Yang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Lingyan Liu
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, China
| | - Xiaojun Zhou
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Shuo Chen
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Lai Wei
- Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Nan Chen
- Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Chuanglong He
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Guang Yang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai 200438, China
| |
Collapse
|
11
|
Ayagama T, Charles PD, Bose SJ, Boland B, Priestman DA, Aston D, Berridge G, Fischer R, Cribbs AP, Song Q, Mirams GR, Amponsah K, Heather L, Galione A, Herring N, Kramer H, Capel RA, Platt FM, Schotten U, Verheule S, Burton RA. Compartmentalization proteomics revealed endolysosomal protein network changes in a goat model of atrial fibrillation. iScience 2024; 27:109609. [PMID: 38827406 PMCID: PMC11141153 DOI: 10.1016/j.isci.2024.109609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/07/2024] [Accepted: 03/25/2024] [Indexed: 06/04/2024] Open
Abstract
Endolysosomes (EL) are known for their role in regulating both intracellular trafficking and proteostasis. EL facilitate the elimination of damaged membranes, protein aggregates, membranous organelles and play an important role in calcium signaling. The specific role of EL in cardiac atrial fibrillation (AF) is not well understood. We isolated atrial EL organelles from AF goat biopsies and conducted a comprehensive integrated omics analysis to study the EL-specific proteins and pathways. We also performed electron tomography, protein and enzyme assays on these biopsies. Our results revealed the upregulation of the AMPK pathway and the expression of EL-specific proteins that were not found in whole tissue lysates, including GAA, DYNLRB1, CLTB, SIRT3, CCT2, and muscle-specific HSPB2. We also observed structural anomalies, such as autophagic-vacuole formation, irregularly shaped mitochondria, and glycogen deposition. Our results provide molecular information suggesting EL play a role in AF disease process over extended time frames.
Collapse
Affiliation(s)
- Thamali Ayagama
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Samuel J. Bose
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Barry Boland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | | | - Daniel Aston
- Department of Anaesthesia and Critical Care, Royal Papworth Hospital NHS Foundation Trust, Papworth Road, Cambridge CB2 0AY, UK
| | | | - Roman Fischer
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - Adam P. Cribbs
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Headington OX3 7LD, UK
| | - Qianqian Song
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Gary R. Mirams
- Centre for Mathematical Medicine & Biology, Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Kwabena Amponsah
- Centre for Mathematical Medicine & Biology, Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Lisa Heather
- Department of Physiology, Anatomy and Genetics, , University of Oxford, South Park Road, Oxford OX1 3PT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Neil Herring
- Department of Physiology, Anatomy and Genetics, , University of Oxford, South Park Road, Oxford OX1 3PT, UK
| | - Holger Kramer
- Mass spectrometry Facility, The MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | | | | | - Ulrich Schotten
- Departments of Physiology and Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Sander Verheule
- Departments of Physiology and Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rebecca A.B. Burton
- Department of Pharmacology, University of Oxford, Oxford, UK
- University of Liverpool, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| |
Collapse
|
12
|
Bauer J, Vlcek J, Pauly V, Hesse N, Xia R, Mo L, Chivukula AS, Villgrater H, Dressler M, Hildebrand B, Wolf E, Rizas KD, Bauer A, Kääb S, Tomsits P, Schüttler D, Clauss S. Biomarker Periodic Repolarization Dynamics Indicates Enhanced Risk for Arrhythmias and Sudden Cardiac Death in Myocardial Infarction in Pigs. J Am Heart Assoc 2024; 13:e032405. [PMID: 38639363 PMCID: PMC11179938 DOI: 10.1161/jaha.123.032405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/08/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Periodic repolarization dynamics (PRD) is an electrocardiographic biomarker that captures repolarization instability in the low frequency spectrum and is believed to estimate the sympathetic effect on the ventricular myocardium. High PRD indicates an increased risk for postischemic sudden cardiac death (SCD). However, a direct link between PRD and proarrhythmogenic autonomic remodeling has not yet been shown. METHODS AND RESULTS We investigated autonomic remodeling in pigs with myocardial infarction (MI)-related ischemic heart failure induced by balloon occlusion of the left anterior descending artery (n=17) compared with pigs without MI (n=11). Thirty days after MI, pigs demonstrated enhanced sympathetic innervation in the infarct area, border zone, and remote left ventricle paralleled by altered expression of autonomic marker genes/proteins. PRD was enhanced 30 days after MI compared with baseline (pre-MI versus post-MI: 1.75±0.30 deg2 versus 3.29±0.79 deg2, P<0.05) reflecting pronounced autonomic alterations on the level of the ventricular myocardium. Pigs with MI-related ventricular fibrillation and SCD had significantly higher pre-MI PRD than pigs without tachyarrhythmias, suggesting a potential role for PRD as a predictive biomarker for ischemia-related arrhythmias (no ventricular fibrillation versus ventricular fibrillation: 1.50±0.39 deg2 versus 3.18±0.53 deg2 [P<0.05]; no SCD versus SCD: 1.67±0.32 deg2 versus 3.91±0.63 deg2 [P<0.01]). CONCLUSIONS We demonstrate that ischemic heart failure leads to significant proarrhythmogenic autonomic remodeling. The concomitant elevation of PRD levels in pigs with ischemic heart failure and pigs with MI-related ventricular fibrillation/SCD suggests PRD as a biomarker for autonomic remodeling and as a potential predictive biomarker for ventricular arrhythmias/survival in the context of MI.
Collapse
Affiliation(s)
- Julia Bauer
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Julia Vlcek
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Valerie Pauly
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Nora Hesse
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Ruibing Xia
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Li Mo
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Aparna Sharma Chivukula
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Hannes Villgrater
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Marie Dressler
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Bianca Hildebrand
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU MunichMunichGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU MunichMunichGermany
| | - Konstantinos D. Rizas
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
| | - Axel Bauer
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- University Hospital for Internal Medicine IIIMedical University of InnsbruckInnsbruckAustria
| | - Stefan Kääb
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU MunichMunichGermany
| | - Philipp Tomsits
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Dominik Schüttler
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Sebastian Clauss
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU MunichMunichGermany
| |
Collapse
|
13
|
Chen J, Qin H, Hao J, Wang Q, Chen S, Yang G, Li M, Zhu X, Wang D, Chen H, Cui C, Chen M. Cardiac-specific overexpression of CREM-IbΔC-X via CRISPR/Cas9 in mice presents a new model of atrial cardiomyopathy with spontaneous atrial fibrillation. Transl Res 2024; 267:54-66. [PMID: 38199433 DOI: 10.1016/j.trsl.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/13/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Atrial cardiomyopathy (ACM) forms the substrate for atrial fibrillation (AF) and underlies the potential for atrial thrombus formation and subsequent stroke. However, generating stable animal models that accurately replicate the entire progression of atrial lesions, particularly the onset of AF, presents significant challenges. In the present study, we found that the isoform of CRE-binding protein modulator (CREM-IbΔC-X), which is involved in the regulation of cardiac development and atrial rhythm, was highly expressed in atrial biopsies from patients with AF. Building upon this finding, we employed CRISPR/Cas9 technology to create a mouse model with cardiac-specific overexpression of CREM-IbΔC-X (referred to as CS-CREM mice). This animal model effectively illustrated the development of ACM through electrophysiological and structural remodelings over time. Proteomics and Chip-qPCR analysis of atrial samples revealed significant upregulation of cell-matrix adhesion and extracellular matrix structural components, alongside significant downregulation of genes related to atrial functions in the CS-CREM mice. Furthermore, the corresponding responses to anti-arrhythmia drugs, i.e., amiodarone and propafenone, suggested that CS-CREM mice could serve as an ideal in vivo model for drug testing. Our study introduced a novel ACM model with spontaneous AF by cardiac-specifically overexpressing CREM-IbΔC-X in mice, providing valuable insights into the mechanisms and therapeutic targets of ACM.
Collapse
Affiliation(s)
- Jiuzhou Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Huiyuan Qin
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Jingzhe Hao
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Qing Wang
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Shaojie Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Gang Yang
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Mingfang Li
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Xiyu Zhu
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Dongjin Wang
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hongwu Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China.
| | - Chang Cui
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China.
| | - Minglong Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
14
|
Zhao S, Hulsurkar MM, Lahiri SK, Aguilar-Sanchez Y, Munivez E, Müller FU, Jain A, Malovannaya A, Yiu CHK, Reilly S, Wehrens XHT. Atrial proteomic profiling reveals a switch towards profibrotic gene expression program in CREM-IbΔC-X mice with persistent atrial fibrillation. J Mol Cell Cardiol 2024; 190:1-12. [PMID: 38514002 DOI: 10.1016/j.yjmcc.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/19/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Overexpression of the CREM (cAMP response element-binding modulator) isoform CREM-IbΔC-X in transgenic mice (CREM-Tg) causes the age-dependent development of spontaneous AF. PURPOSE To identify key proteome signatures and biological processes accompanying the development of persistent AF through integrated proteomics and bioinformatics analysis. METHODS Atrial tissue samples from three CREM-Tg mice and three wild-type littermates were subjected to unbiased mass spectrometry-based quantitative proteomics, differential expression and pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. RESULTS A total of 98 differentially expressed proteins were identified. Gene ontology analysis revealed enrichment for biological processes regulating actin cytoskeleton organization and extracellular matrix (ECM) dynamics. Changes in ITGAV, FBLN5, and LCP1 were identified as being relevant to atrial fibrosis and structural based on expression changes, co-expression patterns, and PPI network analysis. Comparative analysis with previously published datasets revealed a shift in protein expression patterns from ion-channel and metabolic regulators in young CREM-Tg mice to profibrotic remodeling factors in older CREM-Tg mice. Furthermore, older CREM-Tg mice exhibited protein expression patterns reminiscent of those seen in humans with persistent AF. CONCLUSIONS This study uncovered distinct temporal changes in atrial protein expression patterns with age in CREM-Tg mice consistent with the progressive evolution of AF. Future studies into the role of the key differentially abundant proteins identified in this study in AF progression may open new therapeutic avenues to control atrial fibrosis and substrate development in AF.
Collapse
Affiliation(s)
- Shuai Zhao
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohit M Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Satadru K Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuriana Aguilar-Sanchez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elda Munivez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA; Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chi Him Kendrick Yiu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA; Center for Space Medicine, Baylor College of Medicine, Houston, USA.
| |
Collapse
|
15
|
Lin AE, Bapat AC, Xiao L, Niroula A, Ye J, Wong WJ, Agrawal M, Farady CJ, Boettcher A, Hergott CB, McConkey M, Flores-Bringas P, Shkolnik V, Bick AG, Milan D, Natarajan P, Libby P, Ellinor PT, Ebert BL. Clonal Hematopoiesis of Indeterminate Potential With Loss of Tet2 Enhances Risk for Atrial Fibrillation Through Nlrp3 Inflammasome Activation. Circulation 2024; 149:1419-1434. [PMID: 38357791 PMCID: PMC11058018 DOI: 10.1161/circulationaha.123.065597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP), a common age-associated phenomenon, associates with increased risk of both hematological malignancy and cardiovascular disease. Although CHIP is known to increase the risk of myocardial infarction and heart failure, the influence of CHIP in cardiac arrhythmias, such as atrial fibrillation (AF), is less explored. METHODS CHIP prevalence was determined in the UK Biobank, and incident AF analysis was stratified by CHIP status and clone size using Cox proportional hazard models. Lethally irradiated mice were transplanted with hematopoietic-specific loss of Tet2, hematopoietic-specific loss of Tet2 and Nlrp3, or wild-type control and fed a Western diet, compounded with or without NLRP3 (NLR [NACHT, LRR {leucine rich repeat}] family pyrin domain containing protein 3) inhibitor, NP3-361, for 6 to 9 weeks. Mice underwent in vivo invasive electrophysiology studies and ex vivo optical mapping. Cardiomyocytes from Ldlr-/- mice with hematopoietic-specific loss of Tet2 or wild-type control and fed a Western diet were isolated to evaluate calcium signaling dynamics and analysis. Cocultures of pluripotent stem cell-derived atrial cardiomyocytes were incubated with Tet2-deficient bone marrow-derived macrophages, wild-type control, or cytokines IL-1β (interleukin 1β) or IL-6 (interleukin 6). RESULTS Analysis of the UK Biobank showed individuals with CHIP, in particular TET2 CHIP, have increased incident AF. Hematopoietic-specific inactivation of Tet2 increases AF propensity in atherogenic and nonatherogenic mouse models and is associated with increased Nlrp3 expression and CaMKII (Ca2+/calmodulin-dependent protein kinase II) activation, with AF susceptibility prevented by inactivation of Nlrp3. Cardiomyocytes isolated from Ldlr-/- mice with hematopoietic inactivation of Tet2 and fed a Western diet have impaired calcium release from the sarcoplasmic reticulum into the cytosol, contributing to atrial arrhythmogenesis. Abnormal sarcoplasmic reticulum calcium release was recapitulated in cocultures of cardiomyocytes with the addition of Tet2-deficient macrophages or cytokines IL-1β or IL-6. CONCLUSIONS We identified a modest association between CHIP, particularly TET2 CHIP, and incident AF in the UK Biobank population. In a mouse model of AF resulting from hematopoietic-specific inactivation of Tet2, we propose altered calcium handling as an arrhythmogenic mechanism, dependent on Nlrp3 inflammasome activation. Our data are in keeping with previous studies of CHIP in cardiovascular disease, and further studies into the therapeutic potential of NLRP3 inhibition for individuals with TET2 CHIP may be warranted.
Collapse
Affiliation(s)
- Amy Erica Lin
- Division of Cardiovascular Medicine, Department of Medicine (A.E.L., P.L.), Brigham and Women’s Hospital, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Aneesh C. Bapat
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Demoulas Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Department of Medicine (A.C.B., P.T.E.), Massachusetts General Hospital, Boston
| | - Ling Xiao
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Abhishek Niroula
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
- Department of Laboratory Medicine, Lund University, Sweden (A.N.)
- Institute of Biomedicine, SciLifeLab, University of Gothenburg, Sweden (A.N.)
| | - Jiangchuan Ye
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Waihay J. Wong
- Department of Pathology (W.J.W., C.B.H.), Brigham and Women’s Hospital, Boston, MA
| | - Mridul Agrawal
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Christopher J. Farady
- Novartis Institutes for BioMedical Research Forum 1, Basel, Switzerland (C.J.F., A.B.)
| | - Andreas Boettcher
- Novartis Institutes for BioMedical Research Forum 1, Basel, Switzerland (C.J.F., A.B.)
| | - Christopher B. Hergott
- Department of Pathology (W.J.W., C.B.H.), Brigham and Women’s Hospital, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Marie McConkey
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Patricio Flores-Bringas
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Veronica Shkolnik
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (A.G.B.)
| | - David Milan
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Leducq Foundation, Boston, MA (D.M.)
| | - Pradeep Natarajan
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine (A.E.L., P.L.), Brigham and Women’s Hospital, Boston, MA
| | - Patrick T. Ellinor
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Demoulas Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Department of Medicine (A.C.B., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
- Howard Hughes Medical Institute, Boston, MA (B.L.E.)
| |
Collapse
|
16
|
Zhang Z, Vlcek J, Pauly V, Hesse N, Bauer J, Chataut KR, Maderspacher F, Volz LS, Buchberger K, Xia R, Hildebrand B, Kääb S, Schüttler D, Tomsits P, Clauss S. Atrial fibrosis heterogeneity is a risk for atrial fibrillation in pigs with ischaemic heart failure. Eur J Clin Invest 2024; 54:e14137. [PMID: 38012826 DOI: 10.1111/eci.14137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/02/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common arrhythmia and is associated with considerable morbidity and mortality. Ischaemic heart failure (IHF) remains one of the most common causes of AF in clinical practice. However, ischaemia-mediated mechanisms leading to AF are still incompletely understood, and thus, current treatment approaches are limited. To improve our understanding of the pathophysiology, we studied a porcine IHF model. METHODS In pigs, IHF was induced by balloon occlusion of the left anterior descending artery for 90 min. After 30 days of reperfusion, invasive haemodynamic measurements and electrophysiological studies were performed. Masson trichrome and immunofluorescence staining were conducted to assess interstitial fibrosis and myofibroblast activation in different heart regions. RESULTS After 30 days of reperfusion, heart failure with significantly reduced ejection fraction (left anterior obique 30°, 34.78 ± 3.29% [IHF] vs. 62.03 ± 2.36% [control], p < .001; anterior-posterior 0°, 29.16 ± 3.61% vs. 59.54 ± 1.09%, p < .01) was observed. These pigs showed a significantly higher susceptibility to AF (33.90% [IHF] vs. 12.98% [control], p < .05). Histological assessment revealed aggravated fibrosis in atrial appendages but not in atrial free walls in IHF pigs (11.13 ± 1.44% vs. 5.99 ± .86%, p < .01 [LAA], 8.28 ± .56% vs. 6.01 ± .35%, p < .01 [RAA]), which was paralleled by enhanced myofibroblast activation (12.09 ± .65% vs. 9.00 ± .94%, p < .05 [LAA], 14.37 ± .60% vs. 10.30 ± 1.41%, p < .05 [RAA]). Correlation analysis indicated that not fibrosis per se but its cross-regional heterogeneous distribution across the left atrium was associated with AF susceptibility (r = .6344, p < .01). CONCLUSION Our results suggest that left atrial cross-regional fibrosis difference rather than overall fibrosis level is associated with IHF-related AF susceptibility, presumably by establishing local conduction disturbances and heterogeneity.
Collapse
Affiliation(s)
- Zhihao Zhang
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Julia Vlcek
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Valerie Pauly
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Nora Hesse
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Julia Bauer
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Kavi Raj Chataut
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Florian Maderspacher
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Lina Sophie Volz
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Katharina Buchberger
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Ruibing Xia
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Bianca Hildebrand
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
| | - Stefan Kääb
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Dominik Schüttler
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Philipp Tomsits
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| |
Collapse
|
17
|
Seibertz F, Voigt N. High-throughput methods for cardiac cellular electrophysiology studies: the road to personalized medicine. Am J Physiol Heart Circ Physiol 2024; 326:H938-H949. [PMID: 38276947 PMCID: PMC11279751 DOI: 10.1152/ajpheart.00599.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Personalized medicine refers to the tailored application of medical treatment at an individual level, considering the specific genotype or phenotype of each patient for targeted therapy. In the context of cardiovascular diseases, implementing personalized medicine is challenging due to the high costs involved and the slow pace of identifying the pathogenicity of genetic variants, deciphering molecular mechanisms of disease, and testing treatment approaches. Scalable cellular models such as human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) serve as useful in vitro tools that reflect individual patient genetics and retain clinical phenotypes. High-throughput functional assessment of these constructs is necessary to rapidly assess cardiac pathogenicity and test new therapeutics if personalized medicine is to become a reality. High-throughput photometry recordings of single cells coupled with potentiometric probes offer cost-effective alternatives to traditional patch-clamp assessments of cardiomyocyte action potential characteristics. Importantly, automated patch-clamp (APC) is rapidly emerging in the pharmaceutical industry and academia as a powerful method to assess individual membrane-bound ionic currents and ion channel biophysics over multiple cells in parallel. Now amenable to primary cell and hiPSC-CM measurement, APC represents an exciting leap forward in the characterization of a multitude of molecular mechanisms that underlie clinical cardiac phenotypes. This review provides a summary of state-of-the-art high-throughput electrophysiological techniques to assess cardiac electrophysiology and an overview of recent works that successfully integrate these methods into basic science research that could potentially facilitate future implementation of personalized medicine at a clinical level.
Collapse
Affiliation(s)
- Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells," Georg-August University Göttingen, Göttingen, Germany
- Nanion Technologies, GmbH, Munich, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells," Georg-August University Göttingen, Göttingen, Germany
| |
Collapse
|
18
|
Suda M, Katsuumi G, Tchkonia T, Kirkland JL, Minamino T. Potential Clinical Implications of Senotherapies for Cardiovascular Disease. Circ J 2024; 88:277-284. [PMID: 37880106 PMCID: PMC10922738 DOI: 10.1253/circj.cj-23-0657] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Aging is a major risk factor for cardiovascular diseases (CVDs) and accumulating evidence indicates that biological aging has a significant effect on the onset and progression of CVDs. In recent years, therapies targeting senescent cells (senotherapies), particularly senolytics that selectively eliminate senescent cells, have been developed and show promise for treating geriatric syndromes and age-associated diseases, including CVDs. In 2 pilot studies published in 2019 the senolytic combination, dasatinib plus quercetin, improved physical function in patients with idiopathic pulmonary fibrosis and eliminated senescent cells from adipose tissue in patients with diabetic kidney disease. More than 30 clinical trials using senolytics are currently underway or planned. In preclinical CVD models, senolytics appear to improve heart failure, ischemic heart disease, valvular heart disease, atherosclerosis, aortic aneurysm, vascular dysfunction, dialysis arteriovenous fistula patency, and pre-eclampsia. Because senotherapies are completely different strategies from existing treatment paradigms, they might alleviate diseases for which there are no current effective treatments or they could be used in addition to current therapies to enhance efficacy. Moreover, senotherapies might delay, prevent, alleviate or treat multiple diseases in the elderly and reduce polypharmacy, because senotherapies target fundamental aging mechanisms. We comprehensively summarize the preclinical evidence about senotherapies for CVDs and discuss future prospects for their clinical application.
Collapse
Affiliation(s)
- Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
- Department of Medicine and Physiology and Biomedical Engineering, Mayo Clinic
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Tamar Tchkonia
- Department of Medicine and Physiology and Biomedical Engineering, Mayo Clinic
| | - James L Kirkland
- Department of Medicine and Physiology and Biomedical Engineering, Mayo Clinic
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development
| |
Collapse
|
19
|
Zhao S, Hulsurkar MM, Lahiri SK, Aguilar-Sanchez Y, Munivez E, Müller FU, Jain A, Malovannaya A, Yiu K, Reilly S, Wehrens XH. Atrial Proteomic Profiling Reveals a Switch Towards Profibrotic Gene Expression Program in CREM-IbΔC-X Mice with Persistent Atrial Fibrillation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575097. [PMID: 38260363 PMCID: PMC10802622 DOI: 10.1101/2024.01.10.575097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Overexpression of the CREM (cAMP response element-binding modulator) isoform CREM-IbΔC-X in transgenic mice (CREM-Tg) causes the age-dependent development of spontaneous AF. Purpose To identify key proteome signatures and biological processes accompanying the development of persistent AF through integrated proteomics and bioinformatics analysis. Methods Atrial tissue samples from three CREM-Tg mice and three wild-type littermates were subjected to unbiased mass spectrometry-based quantitative proteomics, differential expression and pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. Results A total of 98 differentially expressed proteins were identified. Gene ontology analysis revealed enrichment for biological processes regulating actin cytoskeleton organization and extracellular matrix (ECM) dynamics. Changes in ITGAV, FBLN5, and LCP1 were identified as being relevant to atrial fibrosis and remodeling based on expression changes, co-expression patterns, and PPI network analysis. Comparative analysis with previously published datasets revealed a shift in protein expression patterns from ion-channel and metabolic regulators in young CREM-Tg mice to profibrotic remodeling factors in older CREM-Tg mice. Furthermore, older CREM-Tg mice exhibited protein expression patterns that resembled those of humans with persistent AF. Conclusions This study uncovered distinct temporal changes in atrial protein expression patterns with age in CREM-Tg mice consistent with the progressive evolution of AF. Future studies into the role of the key differentially abundant proteins identified in this study in AF progression may open new therapeutic avenues to control atrial fibrosis and substrate development in AF.
Collapse
Affiliation(s)
- Shuai Zhao
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohit M. Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Satadru K. Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuriana Aguilar-Sanchez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elda Munivez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
- Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kendrick Yiu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Xander H.T. Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, USA
| |
Collapse
|
20
|
Liu Y, Wang D, Jin Y, Sun G, Lou Q, Wang H, Li W. Costunolide ameliorates angiotensin II-induced atrial inflammation and fibrosis by regulating mitochondrial function and oxidative stress in mice: A possible therapeutic approach for atrial fibrillation. Microvasc Res 2024; 151:104600. [PMID: 37666318 DOI: 10.1016/j.mvr.2023.104600] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
Atrial fibrillation (AF) is a cardiac disease characterized by disordered atrial electrical activity. Atrial inflammation and fibrosis are involved in AF progression. Costunolide (COS) is a sesquiterpene lactone containing anti-inflammatory and anti-fibrotic activities. This study aims to explore the underlying mechanisms by which COS protects against AF. Male C57BL/6 mice (8- to 10-week-old) were infused with angiotensin (Ang) II for 3 weeks. Meanwhile, different doses of COS (COS-L: 10 mg/kg, COS-H: 20 mg/kg) were administered to mice by intragastric treatment. The results showed irregular and rapid heart rates in Ang II-treated mice. Moreover, the levels of inflammatory cytokines and fibrotic factors were elevated in mice. COS triggered a reduction of Ang II-induced inflammation and fibrosis, which conferred a protective effect. Mechanistically, mitochondrial dysfunction with mitochondrial respiration inhibition and aberrant ATP levels were observed after Ang II treatment. Moreover, Ang-II-induced excessive reactive oxygen species caused oxidative stress, which was further aggravated by inhibiting Nrf2 nuclear translocation. Importantly, COS diminished these Ang-II-mediated effects in mice. In conclusion, COS attenuated inflammation and fibrosis in Ang-II-treated mice by alleviating mitochondrial dysfunction and oxidative stress. Our findings represent a potential therapeutic option for AF treatment.
Collapse
Affiliation(s)
- Yushu Liu
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Dong Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, PR China
| | - Yimin Jin
- Department of General Practice, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Guifang Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Qi Lou
- Graduate Student, Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Hong Wang
- Graduate Student, Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Weimin Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China.
| |
Collapse
|
21
|
Fisher SM, Murally AR, Rajabally Z, Almas T, Azhar M, Cheema FH, Malone A, Hasan B, Aslam N, Saidi J, O'Neill J, Hameed A. Large animal models to study effectiveness of therapy devices in the treatment of heart failure with preserved ejection fraction (HFpEF). Heart Fail Rev 2024; 29:257-276. [PMID: 37999821 DOI: 10.1007/s10741-023-10371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 11/25/2023]
Abstract
Our understanding of the complex pathophysiology of Heart failure with preserved ejection fraction (HFpEF) is limited by the lack of a robust in vivo model. Existing in-vivo models attempt to reproduce the four main phenotypes of HFpEF; ageing, obesity, diabetes mellitus and hypertension. To date, there is no in vivo model that represents all the haemodynamic characteristics of HFpEF, and only a few have proven to be reliable for the preclinical evaluation of potentially new therapeutic targets. HFpEF accounts for 50% of all the heart failure cases and its incidence is on the rise, posing a huge economic burden on the health system. Patients with HFpEF have limited therapeutic options available. The inadequate effectiveness of current pharmaceutical therapeutics for HFpEF has prompted the development of device-based treatments that target the hemodynamic changes to reduce the symptoms of HFpEF. However, despite the potential of device-based solutions to treat HFpEF, most of these therapies are still in the developmental stage and a relevant HFpEF in vivo model will surely expedite their development process. This review article outlines the major limitations of the current large in-vivo models in use while discussing how these designs have helped in the development of therapy devices for the treatment of HFpEF.
Collapse
Affiliation(s)
- Shane Michael Fisher
- Health Sciences Centre, UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Dublin, Ireland
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Anjali Rosanna Murally
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
- School of Medicine, RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Zahra Rajabally
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
- School of Medicine, RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Talal Almas
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Maimoona Azhar
- Graduate Entry Medicine, School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2, 123 St. Stephen's Green, Dublin, D02 YN77, Ireland
| | - Faisal H Cheema
- Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX, USA
| | - Andrew Malone
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Babar Hasan
- Division of Cardiothoracic Sciences, Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Nadeem Aslam
- Division of Cardiothoracic Sciences, Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Jemil Saidi
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Jim O'Neill
- Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland.
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland - RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Dublin, D02 YN77, Ireland.
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.
| |
Collapse
|
22
|
Seibertz F, Rubio T, Springer R, Popp F, Ritter M, Liutkute A, Bartelt L, Stelzer L, Haghighi F, Pietras J, Windel H, Pedrosa NDI, Rapedius M, Doering Y, Solano R, Hindmarsh R, Shi R, Tiburcy M, Bruegmann T, Kutschka I, Streckfuss-Bömeke K, Kensah G, Cyganek L, Zimmermann WH, Voigt N. Atrial fibrillation-associated electrical remodelling in human induced pluripotent stem cell-derived atrial cardiomyocytes: a novel pathway for antiarrhythmic therapy development. Cardiovasc Res 2023; 119:2623-2637. [PMID: 37677054 PMCID: PMC10730244 DOI: 10.1093/cvr/cvad143] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023] Open
Abstract
AIMS Atrial fibrillation (AF) is associated with tachycardia-induced cellular electrophysiology alterations which promote AF chronification and treatment resistance. Development of novel antiarrhythmic therapies is hampered by the absence of scalable experimental human models that reflect AF-associated electrical remodelling. Therefore, we aimed to assess if AF-associated remodelling of cellular electrophysiology can be simulated in human atrial-like cardiomyocytes derived from induced pluripotent stem cells in the presence of retinoic acid (iPSC-aCM), and atrial-engineered human myocardium (aEHM) under short term (24 h) and chronic (7 days) tachypacing (TP). METHODS AND RESULTS First, 24-h electrical pacing at 3 Hz was used to investigate whether AF-associated remodelling in iPSC-aCM and aEHM would ensue. Compared to controls (24 h, 1 Hz pacing) TP-stimulated iPSC-aCM presented classical hallmarks of AF-associated remodelling: (i) decreased L-type Ca2+ current (ICa,L) and (ii) impaired activation of acetylcholine-activated inward-rectifier K+ current (IK,ACh). This resulted in action potential shortening and an absent response to the M-receptor agonist carbachol in both iPSC-aCM and aEHM subjected to TP. Accordingly, mRNA expression of the channel-subunit Kir3.4 was reduced. Selective IK,ACh blockade with tertiapin reduced basal inward-rectifier K+ current only in iPSC-aCM subjected to TP, thereby unmasking an agonist-independent constitutively active IK,ACh. To allow for long-term TP, we developed iPSC-aCM and aEHM expressing the light-gated ion-channel f-Chrimson. The same hallmarks of AF-associated remodelling were observed after optical-TP. In addition, continuous TP (7 days) led to (i) increased amplitude of inward-rectifier K+ current (IK1), (ii) hyperpolarization of the resting membrane potential, (iii) increased action potential-amplitude and upstroke velocity as well as (iv) reversibly impaired contractile function in aEHM. CONCLUSIONS Classical hallmarks of AF-associated remodelling were mimicked through TP of iPSC-aCM and aEHM. The use of the ultrafast f-Chrimson depolarizing ion channel allowed us to model the time-dependence of AF-associated remodelling in vitro for the first time. The observation of electrical remodelling with associated reversible contractile dysfunction offers a novel platform for human-centric discovery of antiarrhythmic therapies.
Collapse
Affiliation(s)
- Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
| | - Tony Rubio
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Robin Springer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Fiona Popp
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Melanie Ritter
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Aiste Liutkute
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Lena Bartelt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Lea Stelzer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Fereshteh Haghighi
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Jan Pietras
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Hendrik Windel
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Núria Díaz i Pedrosa
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | | | - Yannic Doering
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Richard Solano
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Robin Hindmarsh
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
| | - Runzhu Shi
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Tobias Bruegmann
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Ingo Kutschka
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - George Kensah
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany
- Campus-Institute Data Science (CIDAS), University of Göttingen, Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
23
|
Hu Z, Ding L, Yao Y. Atrial fibrillation: mechanism and clinical management. Chin Med J (Engl) 2023; 136:2668-2676. [PMID: 37914663 PMCID: PMC10684204 DOI: 10.1097/cm9.0000000000002906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 11/03/2023] Open
Abstract
ABSTRACT Atrial fibrillation (AF), the most common sustained arrhythmia, is associated with a range of symptoms, including palpitations, cognitive impairment, systemic embolism, and increased mortality. It places a significant burden on healthcare systems worldwide. Despite decades of research, the precise mechanisms underlying AF remain elusive. Current understanding suggests that factors like stretch-induced fibrosis, epicardial adipose tissue (EAT), chronic inflammation, autonomic nervous system (ANS) imbalances, and genetic mutations all play significant roles in its development. In recent years, the advent of wearable devices has revolutionized AF diagnosis, enabling timely detection and monitoring. However, balancing early diagnosis with efficient resource utilization presents new challenges for healthcare providers. AF management primarily focuses on stroke prevention and symptom alleviation. Patients at high risk of thromboembolism require anticoagulation therapy, and emerging pipeline drugs, particularly factor XI inhibitors, hold promise for achieving effective anticoagulation with reduced bleeding risks. The scope of indications for catheter ablation in AF has expanded significantly. Pulsed field ablation, as a novel energy source, shows potential for improving success rates while ensuring safety. This review integrates existing knowledge and ongoing research on AF pathophysiology and clinical management, with emphasis on diagnostic devices, next-generation anticoagulants, drugs targeting underlying mechanisms, and interventional therapies. It offers a comprehensive mosaic of AF, providing insights into its complexities.
Collapse
Affiliation(s)
| | | | - Yan Yao
- Cardiac Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
24
|
Zhou Y, Sun C, Ma Y, Huang Y, Wu K, Huang S, Lin Q, Zhu J, Ning Z, Liu N, Tu T, Liu Q. Identification and validation of aging-related genes in atrial fibrillation. PLoS One 2023; 18:e0294282. [PMID: 37956134 PMCID: PMC10642816 DOI: 10.1371/journal.pone.0294282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in the clinic. Aging plays an essential role in the occurrence and development of AF. Herein, we aimed to identify the aging-related genes associated with AF using bioinformatics analysis. Transcriptome profiles of AF were obtained from the GEO database. Differential expression analysis was performed to identify AF-specific aging-related genes. GO and KEGG enrichment analyses were performed. Subsequently, the LASSO, SVM-RFE, and MCC algorithms were applied to screen aging-related genes. The mRNA expression of the screened genes was validated in the left atrial samples of aged rapid atrial pacing-induced AF canine models and their counterparts. The ROC curves of them were drawn to evaluate their diagnostic potential. Moreover, CIBERSORT was used to estimate immune infiltration. A correlation analysis between screened aging-related genes and infiltrating immune cells was performed. A total of 24 aging-related genes were identified, which were found to be mainly involved in the FoxO signaling pathway, PI3K-Akt signaling pathway, longevity regulating pathway, and peroxisome according to functional enrichment analysis. LASSO, SVM-RFE, and MCC algorithms identified three genes (HSPA9, SOD2, TXN). Furthermore, the expression levels of HSPA9 and SOD2 were validated in aged rapid atrial pacing-induced AF canine models. HSPA9 and SOD2 could be potential diagnostic biomarkers for AF, as evidenced by the ROC curves. Immune infiltration and correlation analysis revealed that HSPA9 and SOD2 were related to immune cell infiltrates. Collectively, these findings provide novel insights into the potential aging-related genes associated with AF. HSPA9 and SOD2 may play a significant role in the occurrence and development of AF.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Chao Sun
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Yunyin Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Shengyuan Huang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Qiuzhen Lin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Jiayi Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Zuodong Ning
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Ningyuan Liu
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Tao Tu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| |
Collapse
|
25
|
Sharma AK, Singh S, Bhat M, Gill K, Zaid M, Kumar S, Shakya A, Tantray J, Jose D, Gupta R, Yangzom T, Sharma RK, Sahu SK, Rathore G, Chandolia P, Singh M, Mishra A, Raj S, Gupta A, Agarwal M, Kifayat S, Gupta A, Gupta P, Vashist A, Vaibhav P, Kathuria N, Yadav V, Singh RP, Garg A. New drug discovery of cardiac anti-arrhythmic drugs: insights in animal models. Sci Rep 2023; 13:16420. [PMID: 37775650 PMCID: PMC10541452 DOI: 10.1038/s41598-023-41942-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/04/2023] [Indexed: 10/01/2023] Open
Abstract
Cardiac rhythm regulated by micro-macroscopic structures of heart. Pacemaker abnormalities or disruptions in electrical conduction, lead to arrhythmic disorders may be benign, typical, threatening, ultimately fatal, occurs in clinical practice, patients on digitalis, anaesthesia or acute myocardial infarction. Both traditional and genetic animal models are: In-vitro: Isolated ventricular Myocytes, Guinea pig papillary muscles, Patch-Clamp Experiments, Porcine Atrial Myocytes, Guinea pig ventricular myocytes, Guinea pig papillary muscle: action potential and refractory period, Langendorff technique, Arrhythmia by acetylcholine or potassium. Acquired arrhythmia disorders: Transverse Aortic Constriction, Myocardial Ischemia, Complete Heart Block and AV Node Ablation, Chronic Tachypacing, Inflammation, Metabolic and Drug-Induced Arrhythmia. In-Vivo: Chemically induced arrhythmia: Aconitine antagonism, Digoxin-induced arrhythmia, Strophanthin/ouabain-induced arrhythmia, Adrenaline-induced arrhythmia, and Calcium-induced arrhythmia. Electrically induced arrhythmia: Ventricular fibrillation electrical threshold, Arrhythmia through programmed electrical stimulation, sudden coronary death in dogs, Exercise ventricular fibrillation. Genetic Arrhythmia: Channelopathies, Calcium Release Deficiency Syndrome, Long QT Syndrome, Short QT Syndrome, Brugada Syndrome. Genetic with Structural Heart Disease: Arrhythmogenic Right Ventricular Cardiomyopathy/Dysplasia, Dilated Cardiomyopathy, Hypertrophic Cardiomyopathy, Atrial Fibrillation, Sick Sinus Syndrome, Atrioventricular Block, Preexcitation Syndrome. Arrhythmia in Pluripotent Stem Cell Cardiomyocytes. Conclusion: Both traditional and genetic, experimental models of cardiac arrhythmias' characteristics and significance help in development of new antiarrhythmic drugs.
Collapse
Affiliation(s)
- Ashish Kumar Sharma
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India.
| | - Shivam Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mehvish Bhat
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Kartik Gill
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mohammad Zaid
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anjali Shakya
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Junaid Tantray
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Divyamol Jose
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Rashmi Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Tsering Yangzom
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Rajesh Kumar Sharma
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | | | - Gulshan Rathore
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Priyanka Chandolia
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mithilesh Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anurag Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Shobhit Raj
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Archita Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mohit Agarwal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Sumaiya Kifayat
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anamika Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Prashant Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Ankit Vashist
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Parth Vaibhav
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Nancy Kathuria
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Vipin Yadav
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Ravindra Pal Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Arun Garg
- MVN University, Palwal, Haryana, India
| |
Collapse
|
26
|
Huiskes FG, Creemers EE, Brundel BJJM. Dissecting the Molecular Mechanisms Driving Electropathology in Atrial Fibrillation: Deployment of RNA Sequencing and Transcriptomic Analyses. Cells 2023; 12:2242. [PMID: 37759465 PMCID: PMC10526291 DOI: 10.3390/cells12182242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Despite many efforts to treat atrial fibrillation (AF), the most common progressive and age-related cardiac tachyarrhythmia in the Western world, the efficacy is still suboptimal. A plausible reason for this is that current treatments are not directed at underlying molecular root causes that drive electrical conduction disorders and AF (i.e., electropathology). Insights into AF-induced transcriptomic alterations may aid in a deeper understanding of electropathology. Specifically, RNA sequencing (RNA-seq) facilitates transcriptomic analyses and discovery of differences in gene expression profiles between patient groups. In the last decade, various RNA-seq studies have been conducted in atrial tissue samples of patients with AF versus controls in sinus rhythm. Identified differentially expressed molecular pathways so far include pathways related to mechanotransduction, ECM remodeling, ion channel signaling, and structural tissue organization through developmental and inflammatory signaling pathways. In this review, we provide an overview of the available human AF RNA-seq studies and highlight the molecular pathways identified. Additionally, a comparison is made between human RNA-seq findings with findings from experimental AF model systems and we discuss contrasting findings. Finally, we elaborate on new exciting RNA-seq approaches, including single-nucleotide variants, spatial transcriptomics and profiling of different populations of total RNA, small RNA and long non-coding RNA.
Collapse
Affiliation(s)
- Fabries G. Huiskes
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, VUmc, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ, Amsterdam, The Netherlands;
- Department of Experimental Cardiology, Amsterdam UMC, Location AMC, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1105 AZ Amsterdam, The Netherlands;
| | - Esther E. Creemers
- Department of Experimental Cardiology, Amsterdam UMC, Location AMC, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1105 AZ Amsterdam, The Netherlands;
| | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, VUmc, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ, Amsterdam, The Netherlands;
| |
Collapse
|
27
|
Cofiño-Fabres C, Passier R, Schwach V. Towards Improved Human In Vitro Models for Cardiac Arrhythmia: Disease Mechanisms, Treatment, and Models of Atrial Fibrillation. Biomedicines 2023; 11:2355. [PMID: 37760796 PMCID: PMC10525681 DOI: 10.3390/biomedicines11092355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Heart rhythm disorders, arrhythmias, place a huge economic burden on society and have a large impact on the quality of life of a vast number of people. Arrhythmias can have genetic causes but primarily arise from heart tissue remodeling during aging or heart disease. As current therapies do not address the causes of arrhythmias but only manage the symptoms, it is of paramount importance to generate innovative test models and platforms for gaining knowledge about the underlying disease mechanisms which are compatible with drug screening. In this review, we outline the most important features of atrial fibrillation (AFib), the most common cardiac arrhythmia. We will discuss the epidemiology, risk factors, underlying causes, and present therapies of AFib, as well as the shortcomings and opportunities of current models for cardiac arrhythmia, including animal models, in silico and in vitro models utilizing human pluripotent stem cell (hPSC)-derived cardiomyocytes.
Collapse
Affiliation(s)
- Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands;
| |
Collapse
|
28
|
Baena-Montes JM, Kraśny MJ, O’Halloran M, Dunne E, Quinlan LR. In Vitro Models for Improved Therapeutic Interventions in Atrial Fibrillation. J Pers Med 2023; 13:1237. [PMID: 37623487 PMCID: PMC10455620 DOI: 10.3390/jpm13081237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Atrial fibrillation is the most common type of cardiac arrhythmias in humans, mostly caused by hyper excitation of specific areas in the atrium resulting in dyssynchronous atrial contractions, leading to severe consequences such as heart failure and stroke. Current therapeutics aim to target this condition through both pharmacological and non-pharmacological approaches. To test and validate any of these treatments, an appropriate preclinical model must be carefully chosen to refine and optimise the therapy features to correctly reverse this condition. A broad range of preclinical models have been developed over the years, with specific features and advantages to closely mimic the pathophysiology of atrial fibrillation. In this review, currently available models are described, from traditional animal models and in vitro cell cultures to state-of-the-art organoids and organs-on-a-chip. The advantages, applications and limitations of each model are discussed, providing the information to select the appropriate model for each research application.
Collapse
Affiliation(s)
- Jara M. Baena-Montes
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, Human Biology Building, University of Galway, H91 TK33 Galway, Ireland
| | - Marcin J. Kraśny
- Smart Sensors Lab, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Martin O’Halloran
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
- Electrical & Electronic Engineering, School of Engineering, University of Galway, H91 TK33 Galway, Ireland
| | - Eoghan Dunne
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Leo R. Quinlan
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, Human Biology Building, University of Galway, H91 TK33 Galway, Ireland
- CÚRAM SFI Centre for Research in Medical Devices, University of Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
29
|
Tan Y, Coyle RC, Barrs RW, Silver SE, Li M, Richards DJ, Lin Y, Jiang Y, Wang H, Menick DR, Deleon-Pennell K, Tian B, Mei Y. Nanowired human cardiac organoid transplantation enables highly efficient and effective recovery of infarcted hearts. SCIENCE ADVANCES 2023; 9:eadf2898. [PMID: 37540743 PMCID: PMC10403216 DOI: 10.1126/sciadv.adf2898] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
Human cardiac organoids hold remarkable potential for cardiovascular disease modeling and human pluripotent stem cell-derived cardiomyocyte (hPSC-CM) transplantation. Here, we show cardiac organoids engineered with electrically conductive silicon nanowires (e-SiNWs) significantly enhance the therapeutic efficacy of hPSC-CMs to treat infarcted hearts. We first demonstrated the biocompatibility of e-SiNWs and their capacity to improve cardiac microtissue engraftment in healthy rat myocardium. Nanowired human cardiac organoids were then engineered with hPSC-CMs, nonmyocyte supporting cells, and e-SiNWs. Nonmyocyte supporting cells promoted greater ischemia tolerance of cardiac organoids, and e-SiNWs significantly improved electrical pacing capacity. After transplantation into ischemia/reperfusion-injured rat hearts, nanowired cardiac organoids significantly improved contractile development of engrafted hPSC-CMs, induced potent cardiac functional recovery, and reduced maladaptive left ventricular remodeling. Compared to contemporary studies with an identical injury model, greater functional recovery was achieved with a 20-fold lower dose of hPSC-CMs, revealing therapeutic synergy between conductive nanomaterials and human cardiac organoids for efficient heart repair.
Collapse
Affiliation(s)
- Yu Tan
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Robert C. Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Mei Li
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Dylan J. Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Yiliang Lin
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Yuanwen Jiang
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Donald R. Menick
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kristine Deleon-Pennell
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bozhi Tian
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
30
|
Jameson HS, Hanley A, Hill MC, Xiao L, Ye J, Bapat A, Ronzier E, Hall AW, Hucker WJ, Clauss S, Barazza M, Silber E, Mina J, Tucker NR, Mills RW, Dong JT, Milan DJ, Ellinor PT. Loss of the Atrial Fibrillation-Related Gene, Zfhx3, Results in Atrial Dilation and Arrhythmias. Circ Res 2023; 133:313-329. [PMID: 37449401 PMCID: PMC10527554 DOI: 10.1161/circresaha.123.323029] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND ZFHX3 (zinc finger homeobox 3), a gene that encodes a large transcription factor, is at the second-most significantly associated locus with atrial fibrillation (AF), but its function in the heart is unknown. This study aims to identify causative genetic variation related to AF at the ZFHX3 locus and examine the impact of Zfhx3 loss on cardiac function in mice. METHODS CRISPR-Cas9 genome editing, chromatin immunoprecipitation, and luciferase assays in pluripotent stem cell-derived cardiomyocytes were used to identify causative genetic variation related to AF at the ZFHX3 locus. Cardiac function was assessed by echocardiography, magnetic resonance imaging, electrophysiology studies, calcium imaging, and RNA sequencing in mice with heterozygous and homozygous cardiomyocyte-restricted Zfhx3 loss (Zfhx3 Het and knockout, respectively). Human cardiac single-nucleus ATAC (assay for transposase-accessible chromatin)-sequencing data was analyzed to determine which genes in atrial cardiomyocytes are directly regulated by ZFHX3. RESULTS We found single-nucleotide polymorphism (SNP) rs12931021 modulates an enhancer regulating ZFHX3 expression, and the AF risk allele is associated with decreased ZFHX3 transcription. We observed a gene-dose response in AF susceptibility with Zfhx3 knockout mice having higher incidence, frequency, and burden of AF than Zfhx3 Het and wild-type mice, with alterations in conduction velocity, atrial action potential duration, calcium handling and the development of atrial enlargement and thrombus, and dilated cardiomyopathy. Zfhx3 loss results in atrial-specific differential effects on genes and signaling pathways involved in cardiac pathophysiology and AF. CONCLUSIONS Our findings implicate ZFHX3 as the causative gene at the 16q22 locus for AF, and cardiac abnormalities caused by loss of cardiac Zfhx3 are due to atrial-specific dysregulation of pathways involved in AF susceptibility. Together, these data reveal a novel and important role for Zfhx3 in the control of cardiac genes and signaling pathways essential for normal atrial function.
Collapse
Affiliation(s)
- Heather S. Jameson
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Alan Hanley
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew C. Hill
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ling Xiao
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Jiangchuan Ye
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Aneesh Bapat
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Elsa Ronzier
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Amelia Weber Hall
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - William J. Hucker
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Sebastian Clauss
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine I, University Hospital Munich, Campus Grosshadern, Ludwig-Maximilians University Munich (LMU), 81377 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Munich, Germany
- Walter Brendel Centre of Experimental Medicine, University Hospital, LMU Munich, Germany
| | - Miranda Barazza
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth Silber
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Julie Mina
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Robert W. Mills
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
31
|
Lee S, Khrestian C, Laurita D, Juzbasich D, Wallick D, Waldo A. Validation of a new species for studying postoperative atrial fibrillation: Swine sterile pericarditis model. Pacing Clin Electrophysiol 2023; 46:1003-1009. [PMID: 37377345 DOI: 10.1111/pace.14765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/02/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND The canine sterile pericarditis model associated with atrial inflammation is an experimental counterpart of postoperative atrial fibrillation (POAF). However, the use of canines for research is restricted by ethics committees in many countries, and social acceptance is declining. OBJECTIVE To validate the feasibility of the swine sterile pericarditis model as an experimental counterpart to study POAF. METHODS Seven domestic pigs (35-60 kg) underwent initial pericarditis surgery. On two or more postoperative days in the closed-chest state, we performed electrophysiological measurements of pacing threshold and atrial effective refractory period (AERP) while pacing from the right atrial appendage (RAA) and the posterior left atrium (PLA). The inducibility of POAF (>5 min) by burst pacing was determined in both the conscious and anesthetized closed-chest state. These data were compared to previously published canine sterile pericarditis data for validation. RESULTS The pacing threshold increased from day 1 to day 3 (2 ± 0.1 to 3.3 ± 0.6 mA in the RAA, 2.5 ± 0.1 to 4.8 ± 0.2 mA in the PLA). Also, the AERP increased from day 1 to day 3 (118 ± 8 to 157 ± 16 ms in the RAA; 98 ± 4 to 124 ± 2 ms in the PLA, both p < .05). Induction of sustained POAF occurred in 43% (POAF CL range 74-124 ms). All electrophysiologic data from the swine model were consistent with the canine model with respect to (1) the range of both pacing threshold and AERP; (2) the progressive increase in threshold and AERP over time; (3) a 40%-50% incidence of POAF. CONCLUSION A newly developed swine sterile pericarditis model demonstrated electrophysiologic properties consistent with the canine model and patients after open heart surgery.
Collapse
Affiliation(s)
- Seungyup Lee
- Departments of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Celeen Khrestian
- Departments of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Daniel Laurita
- Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dragan Juzbasich
- Departments of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Don Wallick
- Departments of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Albert Waldo
- Departments of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
32
|
Thorpe J, Perry MD, Contreras O, Hurley E, Parker G, Harvey RP, Hill AP, Vandenberg JI. Development of a robust induced pluripotent stem cell atrial cardiomyocyte differentiation protocol to model atrial arrhythmia. Stem Cell Res Ther 2023; 14:183. [PMID: 37501071 PMCID: PMC10373292 DOI: 10.1186/s13287-023-03405-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Atrial fibrillation is the most common arrhythmia syndrome and causes significant morbidity and mortality. Current therapeutics, however, have limited efficacy. Notably, many therapeutics shown to be efficacious in animal models have not proved effective in humans. Thus, there is a need for a drug screening platform based on human tissue. The aim of this study was to develop a robust protocol for generating atrial cardiomyocytes from human-induced pluripotent stem cells. METHODS A novel protocol for atrial differentiation, with optimized timing of retinoic acid during mesoderm formation, was compared to two previously published methods. Each differentiation method was assessed for successful formation of a contractile syncytium, electrical properties assayed by optical action potential recordings and multi-electrode array electrophysiology, and response to the G-protein-gated potassium channel activator, carbamylcholine. Atrial myocyte monolayers, derived using the new differentiation protocol, were further assessed for cardiomyocyte purity, gene expression, and the ability to form arrhythmic rotors in response to burst pacing. RESULTS Application of retinoic acid at day 1 of mesoderm formation resulted in a robust differentiation of atrial myocytes with contractile syncytium forming in 16/18 differentiations across two cell lines. Atrial-like myocytes produced have shortened action potentials and field potentials, when compared to standard application of retinoic acid at the cardiac mesoderm stage. Day 1 retinoic acid produced atrial cardiomyocytes are also carbamylcholine sensitive, indicative of active Ikach currents, which was distinct from ventricular myocytes and standard retinoic addition in matched differentiations. A current protocol utilizing reduced Activin A and BMP4 can produce atrial cardiomyocytes with equivalent functionality but with reduced robustness of differentiation; only 8/17 differentiations produced a contractile syncytium. The day 1 retinoic acid protocol was successfully applied to 6 iPSC lines (3 male and 3 female) without additional optimization or modification. Atrial myocytes produced could also generate syncytia with rapid conduction velocities, > 40 cm s-1, and form rotor style arrhythmia in response to burst pacing. CONCLUSIONS This method combines an enhanced atrial-like phenotype with robustness of differentiation, which will facilitate further research in human atrial arrhythmia and myopathies, while being economically viable for larger anti-arrhythmic drug screens.
Collapse
Affiliation(s)
- Jordan Thorpe
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Matthew D Perry
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Department of Pharmacology, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Osvaldo Contreras
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Emily Hurley
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - George Parker
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney, NSW, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia.
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia.
| |
Collapse
|
33
|
Kervadec A, Kezos J, Ni H, Yu M, Marchant J, Spiering S, Kannan S, Kwon C, Andersen P, Bodmer R, Grandi E, Ocorr K, Colas AR. Multiplatform modeling of atrial fibrillation identifies phospholamban as a central regulator of cardiac rhythm. Dis Model Mech 2023; 16:dmm049962. [PMID: 37293707 PMCID: PMC10387351 DOI: 10.1242/dmm.049962] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/26/2023] [Indexed: 06/10/2023] Open
Abstract
Atrial fibrillation (AF) is a common and genetically inheritable form of cardiac arrhythmia; however, it is currently not known how these genetic predispositions contribute to the initiation and/or maintenance of AF-associated phenotypes. One major barrier to progress is the lack of experimental systems to investigate the effects of gene function on rhythm parameters in models with human atrial and whole-organ relevance. Here, we assembled a multi-model platform enabling high-throughput characterization of the effects of gene function on action potential duration and rhythm parameters using human induced pluripotent stem cell-derived atrial-like cardiomyocytes and a Drosophila heart model, and validation of the findings using computational models of human adult atrial myocytes and tissue. As proof of concept, we screened 20 AF-associated genes and identified phospholamban loss of function as a top conserved hit that shortens action potential duration and increases the incidence of arrhythmia phenotypes upon stress. Mechanistically, our study reveals that phospholamban regulates rhythm homeostasis by functionally interacting with L-type Ca2+ channels and NCX. In summary, our study illustrates how a multi-model system approach paves the way for the discovery and molecular delineation of gene regulatory networks controlling atrial rhythm with application to AF.
Collapse
Affiliation(s)
- Anaïs Kervadec
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - James Kezos
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Haibo Ni
- Department of Pharmacology, UC Davis, Davis, CA 95616, USA
| | - Michael Yu
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - James Marchant
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sean Spiering
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Suraj Kannan
- Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chulan Kwon
- Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Rolf Bodmer
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | | | - Karen Ocorr
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alexandre R. Colas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
34
|
Calvet C, Seebeck P. What to consider for ECG in mice-with special emphasis on telemetry. Mamm Genome 2023; 34:166-179. [PMID: 36749381 PMCID: PMC10290603 DOI: 10.1007/s00335-023-09977-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 01/16/2023] [Indexed: 02/08/2023]
Abstract
Genetically or surgically altered mice are commonly used as models of human cardiovascular diseases. Electrocardiography (ECG) is the gold standard to assess cardiac electrophysiology as well as to identify cardiac phenotypes and responses to pharmacological and surgical interventions. A variety of methods are used for mouse ECG acquisition under diverse conditions, making it difficult to compare different results. Non-invasive techniques allow only short-term data acquisition and are prone to stress or anesthesia related changes in cardiac activity. Telemetry offers continuous long-term acquisition of ECG data in conscious freely moving mice in their home cage environment. Additionally, it allows acquiring data 24/7 during different activities, can be combined with different challenges and most telemetry systems collect additional physiological parameters simultaneously. However, telemetry transmitters require surgical implantation, the equipment for data acquisition is relatively expensive and analysis of the vast number of ECG data is challenging and time-consuming. This review highlights the limits of non-invasive methods with respect to telemetry. In particular, primary screening using non-invasive methods can give a first hint; however, subtle cardiac phenotypes might be masked or compensated due to anesthesia and stress during these procedures. In addition, we detail the key differences between the mouse and human ECG. It is crucial to consider these differences when analyzing ECG data in order to properly translate the insights gained from murine models to human conditions.
Collapse
Affiliation(s)
- Charlotte Calvet
- Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, Zurich, Switzerland
| | - Petra Seebeck
- Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Keefe JA, Hulsurkar MM, Reilly S, Wehrens XHT. Mouse models of spontaneous atrial fibrillation. Mamm Genome 2023; 34:298-311. [PMID: 36173465 PMCID: PMC10898345 DOI: 10.1007/s00335-022-09964-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in adults, with a prevalence increasing with age. Current clinical management of AF is focused on tertiary prevention (i.e., treating the symptoms and sequelae) rather than addressing the underlying molecular pathophysiology. Robust animal models of AF, particularly those that do not require supraphysiologic stimuli to induce AF (i.e., showing spontaneous AF), enable studies that can uncover the underlying mechanisms of AF. Several mouse models of AF have been described to exhibit spontaneous AF, but pathophysiologic drivers of AF differ among models. Here, we describe relevant AF mechanisms and provide an overview of large and small animal models of AF. We then provide an in-depth review of the spontaneous mouse models of AF, highlighting the relevant AF mechanisms for each model.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mohit M Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
36
|
Murninkas M, Gillis R, Elyagon S, Levi O, Mulla W, Katz A, Etzion Y, Gradwohl G. An objective tool for quantifying atrial fibrillation substrate in rats. Am J Physiol Heart Circ Physiol 2023; 324:H461-H469. [PMID: 36735403 DOI: 10.1152/ajpheart.00728.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The utility of rodents for research related to atrial fibrillation (AF) is growing exponentially. However, the obtained arrhythmic waveforms are often mixed with ventricular signals and the ability to analyze regularity and complexity of such events is limited. Recently, we introduced an implantable quadripolar electrode adapted for advanced atrial electrophysiology in ambulatory rats. Notably, we have found that the implantation itself leads to progressive atrial remodeling, presumably because of mechanical loading of the atria. In the present study, we developed an algorithm to clean the atrial signals from ventricular mixing and thereafter quantify the AF substrate in an objective manner based on waveform complexity. Rats were sequentially examined 1-, 4-, and 8-wk postelectrode implantation using a standard AF triggering protocol. Preburst ventricular mixing was sampled and automatically subtracted based on QRS detection in the ECG. Thereafter, the "pure" atrial signals were analyzed by Lempel-Ziv complexity algorithm and a complexity ratio (CR) was defined for each signal by normalizing the postburst to the preburst values. Receiver operating characteristic (ROC) curve analysis indicated an optimal CR cutoff of 1.236 that detected irregular arrhythmic events with high sensitivity (94.5%), specificity (93.1%), and area under the curve (AUC) (0.96, 95% confidence interval, 0.945-0.976). Automated and unbiased analysis indicated a gradual increase in signal complexity over time with augmentation of high frequencies in power spectrum analysis. Our findings indicate that CR algorithm detects irregularity in a highly efficient manner and can also detect the atrial remodeling induced by electrode implantation. Thus, CR analysis can strongly facilitate standardized AF research in rodents.NEW & NOTEWORTHY Rodents are increasingly used in AF research. However, because of technical difficulties including atrial waveform mixing by ventricular signals, most studies do not discriminate between irregular (i.e., AF) and regular atrial arrhythmias. Here, we develop an unbiased computerized tool to "pure" the atrial signals from ventricular mixing and thereafter analyze AF substrate based on the level of irregularity in an objective manner. This novel tool can facilitate standardized AF research in rodents.
Collapse
Affiliation(s)
- Michael Murninkas
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Roni Gillis
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sigal Elyagon
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Or Levi
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Wesam Mulla
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Amos Katz
- Department of Cardiology, Barzilai Medical Center, Ashkelon and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Gideon Gradwohl
- Medical Engineering Unit, The Jerusalem College of Technology, Jerusalem, Israel
| |
Collapse
|
37
|
Bapat A, Schloss MJ, Yamazoe M, Grune J, Hulsmans M, Milan DJ, Nahrendorf M, Ellinor PT. A Mouse Model of Atrial Fibrillation in Sepsis. Circulation 2023; 147:1047-1049. [PMID: 36972346 PMCID: PMC10057612 DOI: 10.1161/circulationaha.122.060317] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Affiliation(s)
- Aneesh Bapat
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Maximilian J. Schloss
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Division of Cardiac Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Masahiro Yamazoe
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Jana Grune
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Charité, Berlin, Germany
| | - Maarten Hulsmans
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA USA
| |
Collapse
|
38
|
Tomsits P, Volz L, Xia R, Chivukula A, Schüttler D, Clauß S. Medetomidine/midazolam/fentanyl narcosis alters cardiac autonomic tone leading to conduction disorders and arrhythmias in mice. Lab Anim (NY) 2023; 52:85-92. [PMID: 36959339 PMCID: PMC10063441 DOI: 10.1038/s41684-023-01141-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/15/2023] [Indexed: 03/25/2023]
Abstract
Arrhythmias are critical contributors to cardiovascular morbidity and mortality. Therapies are mainly symptomatic and often insufficient, emphasizing the need for basic research to unveil the mechanisms underlying arrhythmias and to enable better and ideally causal therapies. In translational approaches, mice are commonly used to study arrhythmia mechanisms in vivo. Experimental electrophysiology studies in mice are performed under anesthesia with medetomidine/midazolam/fentanyl (MMF) and isoflurane/fentanyl (IF) as commonly used regimens. Despite evidence of adverse effects of individual components on cardiac function, few data are available regarding the specific effects of these regimens on cardiac electrophysiology in mice. Here we present a study investigating the effects of MMF and IF narcosis on cardiac electrophysiology in vivo in C57BL/6N wild-type mice. Telemetry transmitters were implanted in a group of mice, which served as controls for baseline parameters without narcosis. In two other groups of mice, electrocardiogram and invasive electrophysiology studies were performed under narcosis (with either MMF or IF). Basic electrocardiogram parameters, heart rate variability parameters, sinus node and atrioventricular node function, and susceptibility to arrhythmias were assessed. Experimental data suggest a remarkable influence of MMF on cardiac electrophysiology compared with IF and awake animals. While IF only moderately reduced heart rate, MMF led to significant bradycardia, spontaneous arrhythmias, heart rate variability alterations as well as sinus and AV node dysfunction, and increased inducibility of ventricular arrhythmias. On the basis of these observed effects, we suggest avoiding MMF in mice, specifically when studying cardiac electrophysiology, but also whenever a regular heartbeat is required for reliable results, such as in heart failure or imaging research.
Collapse
Affiliation(s)
- Philipp Tomsits
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Campus Grosshadern and Innenstadt, Ludwig-Maximilians University Munich (LMU), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany.
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Lina Volz
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Campus Grosshadern and Innenstadt, Ludwig-Maximilians University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Ruibing Xia
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Campus Grosshadern and Innenstadt, Ludwig-Maximilians University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Aparna Chivukula
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Campus Grosshadern and Innenstadt, Ludwig-Maximilians University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Dominik Schüttler
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Campus Grosshadern and Innenstadt, Ludwig-Maximilians University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Clauß
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Campus Grosshadern and Innenstadt, Ludwig-Maximilians University Munich (LMU), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany.
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany.
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany.
| |
Collapse
|
39
|
Pauly V, Vlcek J, Zhang Z, Hesse N, Xia R, Bauer J, Loy S, Schneider S, Renner S, Wolf E, Kääb S, Schüttler D, Tomsits P, Clauss S. Effects of Sex on the Susceptibility for Atrial Fibrillation in Pigs with Ischemic Heart Failure. Cells 2023; 12:973. [PMID: 37048048 PMCID: PMC10093477 DOI: 10.3390/cells12070973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Atrial fibrillation (AF) is the most prevalent arrhythmia, often caused by myocardial ischemia/infarction (MI). Men have a 1.5× higher prevalence of AF, whereas women show a higher risk for new onset AF after MI. However, the underlying mechanisms of how sex affects AF pathophysiology are largely unknown. In 72 pigs with/without ischemic heart failure (IHF) we investigated the impact of sex on ischemia-induced proarrhythmic atrial remodeling and the susceptibility for AF. Electrocardiogram (ECG) and electrophysiological studies were conducted to assess electrical remodeling; histological analyses were performed to assess atrial fibrosis in male and female pigs. IHF pigs of both sexes showed a significantly increased vulnerability for AF, but in male pigs more and longer episodes were observed. Unchanged conduction properties but enhanced left atrial fibrosis indicated structural rather than electrical remodeling underlying AF susceptibility. Sex differences were only observed in controls with female pigs showing an increased intrinsic heart rate, a prolonged QRS interval and a prolonged sinus node recovery time. In sum, susceptibility for AF is significantly increased both in male and female pigs with ischemic heart failure. Differences between males and females are moderate, including more and longer AF episodes in male pigs and sinus node dysfunction in female pigs.
Collapse
Affiliation(s)
- Valerie Pauly
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Julia Vlcek
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Zhihao Zhang
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Nora Hesse
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Ruibing Xia
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Julia Bauer
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Simone Loy
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Sarah Schneider
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Simone Renner
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Feodor-Lynen-Strasse 19, D-81377 Munich, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Strasse 25, D-81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Hackerstrasse 27, D-85764 Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, D-85764 Neuherberg, Germany
| | - Eckhard Wolf
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Feodor-Lynen-Strasse 19, D-81377 Munich, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Strasse 25, D-81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Hackerstrasse 27, D-85764 Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, D-85764 Neuherberg, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Grosshadern Campus, LMU Munich, Feodor-Lynen-Stasse 25, D-81377 Munich, Germany
| | - Stefan Kääb
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Feodor-Lynen-Strasse 19, D-81377 Munich, Germany
| | - Dominik Schüttler
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Philipp Tomsits
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
| | - Sebastian Clauss
- Grosshadern Campus, Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University (LMU), Marchioninistrasse 15, D-81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, D-81377 Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, LMU Munich, Marchioninistrasse 68, D-81377 Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Feodor-Lynen-Strasse 19, D-81377 Munich, Germany
| |
Collapse
|
40
|
Nakao M, Watanabe M, Miquerol L, Natsui H, Koizumi T, Kadosaka T, Koya T, Hagiwara H, Kamada R, Temma T, de Vries AAF, Anzai T. Optogenetic termination of atrial tachyarrhythmias by brief pulsed light stimulation. J Mol Cell Cardiol 2023; 178:9-21. [PMID: 36965700 DOI: 10.1016/j.yjmcc.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
AIMS The most efficient way to acutely restore sinus rhythm from atrial fibrillation (AF) is electrical cardioversion, which is painful without adequate sedation. Recent studies in various experimental models have indicated that optogenetic termination of AF using light-gated ion channels may provide a myocardium-specific and potentially painless alternative future therapy. However, its underlying mechanism(s) remain(s) incompletely understood. As brief pulsed light stimulation, even without global illumination, can achieve optogenetic AF termination, besides direct conduction block also modulation of action potential (AP) properties may be involved in the termination mechanism. We studied the relationship between optogenetic AP duration (APD) and effective refractory period (ERP) prolongation by brief pulsed light stimulation and termination of atrial tachyarrhythmia (AT). METHODS AND RESULTS Hearts from transgenic mice expressing the H134R variant of channelrhodopsin-2 in atrial myocytes were explanted and perfused retrogradely. AT induced by electrical stimulation was terminated by brief pulsed blue light stimulation (470 nm, 10 ms, 16 mW/mm2) with 68% efficacy. The termination rate was dependent on pulse duration and light intensity. Optogenetically imposed APD and ERP changes were systematically examined and optically monitored. Brief pulsed light stimulation (10 ms, 6 mW/mm2) consistently prolonged APD and ERP when light was applied at different phases of the cardiac action potential. Optical tracing showed light-induced APD prolongation during the termination of AT. CONCLUSION Our results directly demonstrate that cationic channelrhodopsin activation by brief pulsed light stimulation prolongs the atrial refractory period suggesting that this is one of the key mechanisms of optogenetic termination of AT.
Collapse
Affiliation(s)
- Motoki Nakao
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaya Watanabe
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Lucile Miquerol
- Developmental Biology Institute of Marseille, Aix-Marseille Université, CNRS UMR 7288, Campus de Luminy Case 907, CEDEX 9, Marseille 13288, France
| | - Hiroyuki Natsui
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takuya Koizumi
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahide Kadosaka
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Taro Koya
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hikaru Hagiwara
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rui Kamada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Taro Temma
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Antoine A F de Vries
- Laboratory of Experimental Cardiology Department of Cardiology, Leiden University Medical Center Leiden, Netherlands
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
41
|
Moreira JD, Gower AC, Xue L, Alekseyev Y, Smith KK, Choi SH, Ayalon N, Farb MG, Tenan K, LeClerc A, Levy D, Benjamin EJ, Lenburg ME, Mitchell RN, Padera RF, Fetterman JL, Gopal DM. Systematic dissection, preservation, and multiomics in whole human and bovine hearts. Cardiovasc Pathol 2023; 63:107495. [PMID: 36334690 PMCID: PMC10031913 DOI: 10.1016/j.carpath.2022.107495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/22/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVES We sought to develop a rigorous, systematic protocol for the dissection and preservation of human hearts for biobanking that expands previous success in postmortem transcriptomics to multiomics from paired tissue. BACKGROUND Existing cardiac biobanks consist largely of biopsy tissue or explanted hearts in select diseases and are insufficient for correlating whole organ phenotype with clinical data. METHODS We demonstrate optimal conditions for multiomics interrogation (ribonucleic acid (RNA) sequencing, untargeted metabolomics) in hearts by evaluating the effect of technical variables (storage solution, temperature) and simulated postmortem interval (PMI) on RNA and metabolite stability. We used bovine (n=3) and human (n=2) hearts fixed in PAXgene or snap-frozen with liquid nitrogen. RESULTS Using a paired Wald test, only two of the genes assessed were differentially expressed between left ventricular samples from bovine hearts stored in PAXgene at 0 and 12 hours PMI (FDR q<0.05). We obtained similar findings in human left ventricular samples, suggesting stability of RNA transcripts at PMIs up to 12 hours. Different library preparation methods (mRNA poly-A capture vs. rRNA depletion) resulted in similar quality metrics with both library preparations achieving >95% of reads properly aligning to the reference genomes across all PMIs for bovine and human hearts. PMI had no effect on RNA Integrity Number or quantity of RNA recovered at the time points evaluated. Of the metabolites identified (855 total) using untargeted metabolomics of human left ventricular tissue, 503 metabolites remained stable across PMIs (0, 4, 8, 12 hours). Most metabolic pathways retained several stable metabolites. CONCLUSIONS Our data demonstrate a technically rigorous, reproducible protocol that will enhance cardiac biobanking practices and facilitate novel insights into human CVD. CONDENSED ABSTRACT Cardiovascular disease (CVD) is the leading cause of mortality worldwide. Current biobanking practices insufficiently capture both the diverse array of phenotypes present in CVDs and the spatial heterogeneity across cardiac tissue sites. We have developed a rigorous and systematic protocol for the dissection and preservation of human cardiac biospecimens to enhance the availability of whole organ tissue for multiple applications. When combined with longitudinal clinical phenotyping, our protocol will enable multiomics in hearts to deepen our understanding of CVDs.
Collapse
Affiliation(s)
- Jesse D Moreira
- Evans Department of Medicine and The Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Adam C Gower
- Department of Medicine, Section of Computational Biomedicine, and Clinical and Translational Science Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Liying Xue
- Evans Department of Medicine and The Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yuriy Alekseyev
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Karan K Smith
- Evans Department of Medicine and The Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Seung H Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nir Ayalon
- Cardiovascular Medicine Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Melissa G Farb
- Evans Department of Medicine and The Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kenneth Tenan
- BU Microarray and Sequencing Resource, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ashley LeClerc
- BU Microarray and Sequencing Resource, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Department of Medicine, Preventive Medicine & Epidemiology Section, Boston University Chobanian & Avedisian School of Medicine, Boston University and the National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Emelia J Benjamin
- Department of Medicine, Preventive Medicine & Epidemiology Section, Boston University Chobanian & Avedisian School of Medicine, Boston University and the National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA; Section of Cardiovascular Medicine, Boston Medical Center/Boston University Chobanian & Avedisian School of Medicine and Department of Epidemiology Boston University School of Public Health, Boston, MA, USA
| | - Marc E Lenburg
- Department of Medicine, Section of Computational Biomedicine, and Clinical and Translational Science Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Richard N Mitchell
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica L Fetterman
- Evans Department of Medicine and The Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Deepa M Gopal
- Evans Department of Medicine and The Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Cardiovascular Medicine Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
42
|
Godoy-Marín H, Jiménez-Sábado V, Tarifa C, Ginel A, Santos JLD, Bentzen BH, Hove-Madsen L, Ciruela F. Increased Density of Endogenous Adenosine A 2A Receptors in Atrial Fibrillation: From Cellular and Porcine Models to Human Patients. Int J Mol Sci 2023; 24:ijms24043668. [PMID: 36835078 PMCID: PMC9963500 DOI: 10.3390/ijms24043668] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/04/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Adenosine, an endogenous nucleoside, plays a critical role in maintaining homeostasis during stressful situations, such as energy deprivation or cellular damage. Therefore, extracellular adenosine is generated locally in tissues under conditions such as hypoxia, ischemia, or inflammation. In fact, plasma levels of adenosine in patients with atrial fibrillation (AF) are elevated, which also correlates with an increased density of adenosine A2A receptors (A2ARs) both in the right atrium and in peripheral blood mononuclear cells (PBMCs). The complexity of adenosine-mediated effects in health and disease requires simple and reproducible experimental models of AF. Here, we generate two AF models, namely the cardiomyocyte cell line HL-1 submitted to Anemonia toxin II (ATX-II) and a large animal model of AF, the right atrium tachypaced pig (A-TP). We evaluated the density of endogenous A2AR in those AF models. Treatment of HL-1 cells with ATX-II reduced cell viability, while the density of A2AR increased significantly, as previously observed in cardiomyocytes with AF. Next, we generated the animal model of AF based on tachypacing pigs. In particular, the density of the key calcium regulatory protein calsequestrin-2 was reduced in A-TP animals, which is consistent with the atrial remodelling shown in humans suffering from AF. Likewise, the density of A2AR in the atrium of the AF pig model increased significantly, as also shown in the biopsies of the right atrium of subjects with AF. Overall, our findings revealed that these two experimental models of AF mimicked the alterations in A2AR density observed in patients with AF, making them attractive models for studying the adenosinergic system in AF.
Collapse
Affiliation(s)
- Héctor Godoy-Marín
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907 L’Hospitalet de Llobregat, Spain
| | - Verónica Jiménez-Sábado
- Biomedical Research Institute Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Carmen Tarifa
- Biomedical Research Institute Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
- Biomedical Research Institute of Barcelona, IIBB-CSIC, 08036 Barcelona, Spain
| | - Antonino Ginel
- Department Cardiac Surgery, Hospital de la Santa Creu i Sant Pau, 08036 Barcelona, Spain
| | | | - Bo Hjorth Bentzen
- Department of Biomedical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Leif Hove-Madsen
- Biomedical Research Institute Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, 28029 Madrid, Spain
- Biomedical Research Institute of Barcelona, IIBB-CSIC, 08036 Barcelona, Spain
- Correspondence: (L.H.-M.); (F.C.)
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907 L’Hospitalet de Llobregat, Spain
- Correspondence: (L.H.-M.); (F.C.)
| |
Collapse
|
43
|
Regional Differences in Ca 2+ Signaling and Transverse-Tubules across Left Atrium from Adult Sheep. Int J Mol Sci 2023; 24:ijms24032347. [PMID: 36768669 PMCID: PMC9916916 DOI: 10.3390/ijms24032347] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Cardiac excitation-contraction coupling can be different between regions of the heart. Little is known at the atria level, specifically in different regions of the left atrium. This is important given the role of cardiac myocytes from the pulmonary vein sleeves, which are responsible for ectopic activity during atrial fibrillation. In this study, we present a new method to isolate atrial cardiac myocytes from four different regions of the left atrium of a large animal model, sheep, highly relevant to humans. Using collagenase/protease we obtained calcium-tolerant atrial cardiac myocytes from the epicardium, endocardium, free wall and pulmonary vein regions. Calcium transients were slower (time to peak and time to decay) in free wall and pulmonary vein myocytes compared to the epicardium and endocardium. This is associated with lower t-tubule density. Overall, these results suggest regional differences in calcium transient and t-tubule density across left atria, which may play a major role in the genesis of atrial fibrillation.
Collapse
|
44
|
Cui M, Liu Y, Men X, Li T, Liu D, Deng Y. Large animal models in the study of gynecological diseases. Front Cell Dev Biol 2023; 11:1110551. [PMID: 36755972 PMCID: PMC9899856 DOI: 10.3389/fcell.2023.1110551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/12/2023] [Indexed: 01/24/2023] Open
Abstract
Gynecological diseases are a series of diseases caused by abnormalities in the female reproductive organs or breast, which endanger women's fertility and even their lives. Therefore, it is important to investigate the mechanism of occurrence and treatment of gynecological diseases. Animal models are the main objects for people to study the development of diseases and explore treatment options. Large animals, compared to small rodents, have reproductive organs with structural and physiological characteristics closer to those of humans, and are also better suited for long-term serial examinations for gynecological disease studies. This review gives examples of large animal models in gynecological diseases and provides a reference for the selection of animal models for gynecological diseases.
Collapse
Affiliation(s)
- Minghua Cui
- Gynecology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yuehui Liu
- Laboratory Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaoping Men
- Laboratory Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Tao Li
- Department of Acupuncture and Massage, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Da Liu, ; Yongzhi Deng,
| | - Yongzhi Deng
- The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Da Liu, ; Yongzhi Deng,
| |
Collapse
|
45
|
Niort BC, Recalde A, Cros C, Brette F. Critical Link between Calcium Regional Heterogeneity and Atrial Fibrillation Susceptibility in Sheep Left Atria. J Clin Med 2023; 12:jcm12030746. [PMID: 36769395 PMCID: PMC9917890 DOI: 10.3390/jcm12030746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/30/2022] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Atrial fibrillation is the most sustained form of arrhythmia in the human population that leads to important electrophysiological and structural cardiac remodeling as it progresses into a chronic form. Calcium is an established key player of cellular electrophysiology in the heart, yet to date, there is no information that maps calcium signaling across the left atrium. OBJECTIVE The aim of this study is to determine whether calcium signaling is homogenous throughout the different regions of the left atrium. This work tests the hypothesis that differences across the healthy left atrium contribute to a unique, region-dependent calcium cycling and participates in the pro-arrhythmic activity during atrial fibrillation. METHODS An animal model relevant to human cardiac function (the sheep) was used to characterize both the electrical activity and the calcium signaling of three distinct left atrium regions (appendage, free wall and pulmonary veins) in control conditions and after acetylcholine perfusion (5 μM) to induce acute atrial fibrillation. High-resolution dual calcium-voltage optical mapping on the left atria of sheep was performed to explore the spatiotemporal dynamics of calcium signaling in relation to electrophysiological properties. RESULTS Action potential duration (at 80% repolarization) was not significantly different in the three regions of interest for the three pacing sites. In contrast, the time to 50% calcium transient decay was significantly different depending on the region paced and recorded. Acetylcholine perfusion and burst pacing-induced atrial fibrillation when pulmonary veins and appendage regions were paced but not when the free wall region was. Dantrolene (a ryanodine receptor blocker) did not reduce atrial fibrillation susceptibility. CONCLUSION These data provide the first evidence of heterogenous calcium signaling across the healthy left atrium. Such basal regional differences may be exacerbated during the progression of atrial fibrillation and thus play a crucial role in focal arrhythmia initiation without ryanodine receptor gating modification.
Collapse
Affiliation(s)
- Barbara C. Niort
- Centre de Recherche Cardio-Thoracique de Bordeaux (CRCTB), Inserm U1045, Univeristé de Bordeaux, F-33000 Bordeaux, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, F-33600 Pessac, France
| | - Alice Recalde
- Centre de Recherche Cardio-Thoracique de Bordeaux (CRCTB), Inserm U1045, Univeristé de Bordeaux, F-33000 Bordeaux, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, F-33600 Pessac, France
| | - Caroline Cros
- Centre de Recherche Cardio-Thoracique de Bordeaux (CRCTB), Inserm U1045, Univeristé de Bordeaux, F-33000 Bordeaux, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, F-33600 Pessac, France
| | - Fabien Brette
- Centre de Recherche Cardio-Thoracique de Bordeaux (CRCTB), Inserm U1045, Univeristé de Bordeaux, F-33000 Bordeaux, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, F-33600 Pessac, France
- Phymedexp Inserm, CNRS, Université de Montpellier, CHRU, F-34295 Montpellier, France
- Correspondence:
| |
Collapse
|
46
|
Schmidt A, Balitzki J, Grmaca L, Vogel J, Boehme P, Boden K, Hüser J, Truebel H, Mondritzki T. "Digital biomarkers" in preclinical heart failure models - a further step towards improved translational research. Heart Fail Rev 2023; 28:249-260. [PMID: 36001250 PMCID: PMC9902409 DOI: 10.1007/s10741-022-10264-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 02/07/2023]
Abstract
Innovations in the development of novel heart failure therapies are essential to further increase the predictive value of early research findings. Animal models are still playing a pivotal role in 'translational research'. In recent years, the transferability from animal studies has been more and more critically discussed due to persistent high attrition rates in clinical trials. However, there is an increasing trend to implement mobile health devices in preclinical studies. These devices can increase the predictive value of animal models by providing more accurate and translatable data and protect from confounding factors. This review outlines the current prevalence and opportunities of these techniques in preclinical heart failure research studies to accelerate the integration of these important tools. A literature screening for preclinical heart failure studies in large animals implementing telemetry devices over the last decade was performed. Twelve out of 43 publications were included. A variety of different hemodynamic and cardiac parameters can be recorded in conscious state by means of telemetry devices in both, the animal model and the patient. The measurement quality is consistently rated as valid and robust. Mobile health technologies functioning as digital biomarkers represent a more predictive approach compared to the traditionally used invasive measurement techniques, due to the possibility of continuous data collection in the conscious animal. Furthermore, they help to implement the 3R concept (reduction, refinement, replacement) in animal research. Despite this, the use of these techniques in preclinical research has been restrained to date.
Collapse
Affiliation(s)
- Alexander Schmidt
- grid.420044.60000 0004 0374 4101Bayer AG, BAG-PH-RD-RED-TA1-CPM-CPM2, Building 0520, 42096 Wuppertal, Germany ,grid.411327.20000 0001 2176 9917Heinrich-Heine-University, Düsseldorf, Germany
| | - Jakob Balitzki
- grid.420044.60000 0004 0374 4101Bayer AG, BAG-PH-RD-RED-TA1-CPM-CPM2, Building 0520, 42096 Wuppertal, Germany ,grid.10423.340000 0000 9529 9877Hannover Medical School, Hannover, Germany
| | - Ljubica Grmaca
- grid.420044.60000 0004 0374 4101Bayer AG, BAG-PH-RD-RED-TA1-CPM-CPM2, Building 0520, 42096 Wuppertal, Germany ,grid.10253.350000 0004 1936 9756Philipps-University of Marburg, Marburg, Germany
| | - Julia Vogel
- grid.420044.60000 0004 0374 4101Bayer AG, BAG-PH-RD-RED-TA1-CPM-CPM2, Building 0520, 42096 Wuppertal, Germany ,grid.412581.b0000 0000 9024 6397University of Witten/Herdecke, Witten, Germany ,grid.5718.b0000 0001 2187 5445Clinic for Cardiology and Angiology, West-German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Duisburg, Germany
| | - Philip Boehme
- grid.412581.b0000 0000 9024 6397University of Witten/Herdecke, Witten, Germany
| | - Katharina Boden
- grid.412581.b0000 0000 9024 6397University of Witten/Herdecke, Witten, Germany
| | - Jörg Hüser
- grid.420044.60000 0004 0374 4101Bayer AG, BAG-PH-RD-RED-TA1-CPM-CPM2, Building 0520, 42096 Wuppertal, Germany
| | - Hubert Truebel
- grid.412581.b0000 0000 9024 6397University of Witten/Herdecke, Witten, Germany
| | - Thomas Mondritzki
- Bayer AG, BAG-PH-RD-RED-TA1-CPM-CPM2, Building 0520, 42096, Wuppertal, Germany. .,University of Witten/Herdecke, Witten, Germany.
| |
Collapse
|
47
|
Murphy MB, Kannankeril PJ, Murray KT. Overview of programmed electrical stimulation to assess atrial fibrillation susceptibility in mice. Front Physiol 2023; 14:1149023. [PMID: 37113690 PMCID: PMC10126433 DOI: 10.3389/fphys.2023.1149023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common human arrhythmia and is associated with increased risk of stroke, dementia, heart failure, and death. Among several animal models that have been used to investigate the molecular determinants of AF, mouse models have become the most prevalent due to low cost, ease of genetic manipulation, and similarity to human disease. Programmed electrical stimulation (PES) using intracardiac or transesophageal atrial pacing is used to induce AF as most mouse models do not develop spontaneous AF. However, there is a lack of standardized methodology resulting in numerous PES protocols in the literature that differ with respect to multiple parameters, including pacing protocol and duration, stimulus amplitude, pulse width, and even the definition of AF. Given this complexity, the selection of the appropriate atrial pacing protocol for a specific model has been arbitrary. Herein we review the development of intracardiac and transesophageal PES, including commonly used protocols, selected experimental models, and advantages and disadvantages of both techniques. We also emphasize detection of artifactual AF induction due to unintended parasympathetic stimulation, which should be excluded from results. We recommend that the optimal pacing protocol to elicit an AF phenotype should be individualized to the specific model of genetic or acquired risk factors, with an analysis using several definitions of AF as an endpoint.
Collapse
|
48
|
Hertel JN, Jerltorp K, Hansen MEH, Isaksen JL, Sattler SM, Linz B, Chaldoupi SM, Jespersen T, Saljic A, Gang U, Manninger M, Linz D. 3D-electroanatomical mapping of the left atrium and catheter-based pulmonary vein isolation in pigs: A practical guide. Front Cardiovasc Med 2023; 10:1139364. [PMID: 36970354 PMCID: PMC10033609 DOI: 10.3389/fcvm.2023.1139364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/14/2023] [Indexed: 03/29/2023] Open
Abstract
Aim To propose a standardized workflow for 3D-electroanatomical mapping guided pulmonary vein isolation in pigs. Materials and methods Danish female landrace pigs were anaesthetized. Ultrasound-guided puncture of both femoral veins was performed and arterial access for blood pressure measurement established. Fluoroscopy- and intracardiac ultrasound-guided passage of the patent foramen ovale or transseptal puncture was performed. Then, 3D-electroanatomical mapping of the left atrium was conducted using a high-density mapping catheter. After mapping all pulmonary veins, an irrigated radiofrequency ablation catheter was used to perform ostial ablation to achieve electrical pulmonary vein isolation. Entrance- and exit-block were confirmed and re-assessed after a 20-min waiting period. Lastly, animals were sacrificed to perform left atrial anatomical gross examination. Results We present data from 11 consecutive pigs undergoing pulmonary vein isolation. Passage of the fossa ovalis or transseptal puncture was uneventful and successful in all animals. Within the inferior pulmonary trunk 2-4 individual veins as well as 1-2 additional left and right pulmonary veins could be cannulated. Electrical isolation by point-by-point ablation of all targeted veins was successful. However, pitfalls including phrenic nerve capture during ablation, ventricular arrhythmias during antral isolation close to the mitral valve annulus and difficulties in accessing right pulmonary veins were encountered. Conclusion Fluoroscopy- and intracardiac ultrasound-guided transseptal puncture, high-density electroanatomical mapping of all pulmonary veins and complete electrical pulmonary vein isolation can be achieved reproducibly and safely in pigs when using current technologies and a step-by-step approach.
Collapse
Affiliation(s)
- Julie Norup Hertel
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kezia Jerltorp
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Malthe Emil Høtbjerg Hansen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas L. Isaksen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefan Michael Sattler
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Gentofte University Hospital, Hellerup, Denmark
| | - Benedikt Linz
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Emergency Department, Frederiksberg and Bispebjerg Hospital, Copenhagen, Denmark
| | - Sevasti-Maria Chaldoupi
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- West German Heart and Vascular Center, Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Uffe Gang
- Department of Cardiology, Zealand University Hospital Roskilde, Roskilde, Denmark
| | - Martin Manninger
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dominik Linz
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Cardiology, Zealand University Hospital Roskilde, Roskilde, Denmark
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, SA, Australia
- *Correspondence: Dominik Linz,
| |
Collapse
|
49
|
Ripplinger CM, Glukhov AV, Kay MW, Boukens BJ, Chiamvimonvat N, Delisle BP, Fabritz L, Hund TJ, Knollmann BC, Li N, Murray KT, Poelzing S, Quinn TA, Remme CA, Rentschler SL, Rose RA, Posnack NG. Guidelines for assessment of cardiac electrophysiology and arrhythmias in small animals. Am J Physiol Heart Circ Physiol 2022; 323:H1137-H1166. [PMID: 36269644 PMCID: PMC9678409 DOI: 10.1152/ajpheart.00439.2022] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 01/09/2023]
Abstract
Cardiac arrhythmias are a major cause of morbidity and mortality worldwide. Although recent advances in cell-based models, including human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM), are contributing to our understanding of electrophysiology and arrhythmia mechanisms, preclinical animal studies of cardiovascular disease remain a mainstay. Over the past several decades, animal models of cardiovascular disease have advanced our understanding of pathological remodeling, arrhythmia mechanisms, and drug effects and have led to major improvements in pacing and defibrillation therapies. There exist a variety of methodological approaches for the assessment of cardiac electrophysiology and a plethora of parameters may be assessed with each approach. This guidelines article will provide an overview of the strengths and limitations of several common techniques used to assess electrophysiology and arrhythmia mechanisms at the whole animal, whole heart, and tissue level with a focus on small animal models. We also define key electrophysiological parameters that should be assessed, along with their physiological underpinnings, and the best methods with which to assess these parameters.
Collapse
Affiliation(s)
- Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
| | - Alexey V Glukhov
- Department of Medicine, Cardiovascular Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Bastiaan J Boukens
- Department Physiology, University Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Nipavan Chiamvimonvat
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
- Department of Internal Medicine, University of California Davis School of Medicine, Davis, California
- Veterans Affairs Northern California Healthcare System, Mather, California
| | - Brian P Delisle
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Larissa Fabritz
- University Center of Cardiovascular Science, University Heart and Vascular Center, University Hospital Hamburg-Eppendorf with DZHK Hamburg/Kiel/Luebeck, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Thomas J Hund
- Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
- Department of Biomedical Engineering, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Na Li
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Katherine T Murray
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Steven Poelzing
- Virginia Tech Carilon School of Medicine, Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech, Roanoke, Virginia
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Stacey L Rentschler
- Cardiovascular Division, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri
| | - Robert A Rose
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nikki G Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia
- Department of Pediatrics, George Washington University School of Medicine, Washington, District of Columbia
| |
Collapse
|
50
|
Butova X, Myachina T, Simonova R, Kochurova A, Bozhko Y, Arkhipov M, Solovyova O, Kopylova G, Shchepkin D, Khokhlova A. Peculiarities of the Acetylcholine Action on the Contractile Function of Cardiomyocytes from the Left and Right Atria in Rats. Cells 2022; 11:cells11233809. [PMID: 36497067 PMCID: PMC9737865 DOI: 10.3390/cells11233809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Acetylcholine (ACh) is the neurotransmitter of the parasympathetic nervous system that modulates cardiac function, and its high concentrations may induce atrial fibrillation. We compared the ACh action on the mechanical function of single cardiomyocytes from the left atria (LA) and the right atria (RA). We exposed single rat LA and RA cardiomyocytes to 1, 10, and 100 µM ACh for 10-15 min and measured the parameters of sarcomere shortening-relengthening and cytosolic calcium ([Ca2+]i) transients during cell contractions. We also studied the effects of ACh on cardiac myosin function using an in vitro motility assay and analyzed the phosphorylation level of sarcomeric proteins. In LA cardiomyocytes, ACh decreased the time to peak sarcomere shortening, time to 50% relengthening, and time to peak [Ca2+]i transients. In RA cardiomyocytes, ACh affected the time of shortening and relengthening only at 10 µM. In the in vitro motility assay, ACh reduced to a greater extent the sliding velocity of F-actin over myosin from LA cardiomyocytes, which was accompanied by a more pronounced decrease in phosphorylation of the myosin regulatory light chain (RLC) in LA cardiomyocytes than in RA cardiomyocytes. Our findings indicate that ACh plays an important role in modulating the contractile function of LA and RA, provoking more pronounced changes in the time course of sarcomere shortening-relengthening and the kinetics of actin-myosin interaction in LA cardiomyocytes.
Collapse
Affiliation(s)
- Xenia Butova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia
| | - Tatiana Myachina
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia
| | - Raisa Simonova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia
| | - Anastasia Kochurova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia
| | - Yakov Bozhko
- Department of Therapy, Ural State Medical University, Repina Str. 3, 620028 Yekaterinburg, Russia
| | - Michael Arkhipov
- Department of Therapy, Ural State Medical University, Repina Str. 3, 620028 Yekaterinburg, Russia
| | - Olga Solovyova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia
- Institute of Natural Sciences and Mathematics, Ural Federal University, Mira 19, 620002 Yekaterinburg, Russia
| | - Galina Kopylova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia
| | - Daniil Shchepkin
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia
| | - Anastasia Khokhlova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Pervomajskaya Str. 106, 620049 Yekaterinburg, Russia
- Institute of Natural Sciences and Mathematics, Ural Federal University, Mira 19, 620002 Yekaterinburg, Russia
- Correspondence:
| |
Collapse
|